Anti-Inflammatory Nutraceuticals and Chronic Diseases: Subash Chandra Gupta Sahdeo Prasad Bharat B. Aggarwal Editors
Anti-Inflammatory Nutraceuticals and Chronic Diseases: Subash Chandra Gupta Sahdeo Prasad Bharat B. Aggarwal Editors
Subash Chandra Gupta
Sahdeo Prasad
Bharat B. Aggarwal Editors
Anti-inflammatory
Nutraceuticals and
Chronic Diseases
Advances in Experimental Medicine and Biology
Volume 928
Series editors
Irun R. Cohen, Rehovot, Israel
N.S. Abel Lajtha, Orangeburg, USA
Rodolfo Paoletti, Milan, Italy
John D. Lambris, Philadelphia, USA
More information about this series at http://www.springer.com/series/5584
Subash Chandra Gupta Sahdeo Prasad
•
Bharat B. Aggarwal
Editors
Anti-inflammatory
Nutraceuticals and Chronic
Diseases
123
Editors
Subash Chandra Gupta Bharat B. Aggarwal
Banaras Hindu University Inflammation Research Institute
Varanasi, Uttar Pradesh San Diego, CA
India USA
Sahdeo Prasad
University of Texas
MD Anderson Cancer Center
Houston, TX
USA
v
vi Contents
vii
viii About the Editors
Abstract Curcumin, a yellow pigment from the spice turmeric, is used in Indian
and Chinese medicine since ancient times for wide range of diseases. Extensive
scientific research on this molecule performed over the last 3 to 4 decades has
proved its potential as an important pharmacological agent. The antioxidant,
anti-inflammatory, antimicrobial and chemopreventive activities of curcumin have
been extended to explore this molecule against many chronic diseases with
promising results. Further, its multitargeting ability and nontoxic nature to humans
even up to 12 g/day have attracted scientists to explore this as an anticancer agent in
the clinic, which is in different phases of trials. With much more scope to be
investigated and understood, curcumin becomes one of the very few inexpensive
botanical molecules with potent therapeutic abilities.
1.1 Introduction
jaundice, skin and eye infections. As a dietary agent, turmeric is regularly used as a
spice and also as a coloring agent in Indian cuisine. Both turmeric and its active
principle, curcumin, are permitted like other natural pigments and the food additive,
E number for curcumin is E100. Depending on the origin and soil conditions, the
percentage of curcuminoids in turmeric varies from 2 to 9 % of its dry weight. The
word “curcuminoid” refers to a mixture of four polyphenols, such as curcumin,
demethoxycurcumin and bis-demethoxy curcumin and cyclic curcumin. Out of
these, curcumin is nearly 70 % of the total curcuminoids. In addition to these
curcuminoids, turmeric also contains essential oils primarily composed of mono
and sesquiterpenes, like turmerones, turmerol, etc. The strong yellow color of
turmeric is mainly due to curcuminoids.
Historically, the first scientific report on isolation and chemical characteristics of
curcumin was made in 1815 [115], and its molecular formula and chemical
structure was published in 1910 [74]. Its first laboratory synthesis was demonstrated
in 1913 [68], subsequently in 1964 a method for producing curcumin in high yields
was published [80], and much later, its biosynthesis was understood [35]. Curcumin
is extracted commercially from turmeric by solvent extraction with ethanol fol-
lowed by column chromatography. Using high-performance liquid chromatography
coupled with absorption, fluorescence and mass detectors, curcumin can be detected
in nanomolar quantities in food samples and biological tissues [35, 90].
Apart from the ancient medicinal documents, early research report on therapeutic
use of curcumin appears to date back to 1748; however, the first scientific document
for treating human disease was reported in 1937 [79], wherein at least 67 patients
were treated for chronic cholecystitis, using curcunat, which is equivalent of cur-
cumin. In this study, oral administration for 3 weeks showed symptomatic
improvement in all cases and radiologic improvement by cholecystogram in 18
patients. Interestingly, no ill effects were observed even when the treatment con-
tinued for several months. The efficacy of curcumin was attributed to its ability to
cause the emptying of the gallbladder. Later in 1949, the antibacterial activity of
curcumin [96] was established, since then and till 1970s there were very few reports
on its biological activities [98, 106]. Initial research investigations were focused
mostly on antioxidant and antibacterial activity and the first anticancer report in
human participants was undertaken by Kuttan et al. [67]. However, after the report
by Singh and Aggarwal [108], confirming the anti-inflammatory activity of cur-
cumin by suppressing NF-κB activity, the pace of curcumin research has progressed
systematically. With several encouraging results in rodent models, curcumin
attracted researchers all over the world, to be developed as a potent anticancer drug.
As per Pubmed website, (as of October 2015) there are 8247 articles reported with
the word “curcumin” in the title, including 808 reviews and 141 clinical trials, out
of these more than half have appeared in the last 5 years. It is well accepted by the
scientific community that no other botanical molecule is as efficient and as scien-
tifically celebrated as curcumin.
1 Curcumin and Its Role in Chronic Diseases 3
H
O O O O
CH3O OCH3 CH3O OCH3
HO OH HO OH
Keto Enol
Fig. 1.1 Chemical structure of curcumin in keto and enol tautomeric forms
water, but is readily soluble in moderately polar solvents like methanol, acetonitrile,
chloroform, dimethylsulfoxide (DMSO), etc. In aqueous solutions its solubility can
be increased by the addition of surfactants, polymers, lipids and proteins. Because
of the presence of serum albumin, clear curcumin solutions in micromolar con-
centration can be prepared in cell culture medium.
Curcumin has three important functional groups, two o-methoxy phenolic groups,
one enone moiety and an α,β-unsaturated diketone group. Each functional group
has some specific role in crucial biological activity in curcumin. The o-methoxy
phenolic-OH group of curcumin is primarily involved in direct scavenging of
reactive oxygen species (ROS), where it donates an electron or hydrogen atom to
the oxidizing radicals and the resultant curcumin phenoxyl radical acquires stability
through the conjugation and resonating structures [87]. This phenoxyl radical is
regenerated back to curcumin by water-soluble antioxidants like ascorbic acid
making it an excellent chain-breaking antioxidant that has been reported to be at
least ten times better than vitamin E. Curcumin binds to many biomolecules like
proteins, lipids and DNA [42, 88]. The proteins that interact with curcumin include
transcription factors, inflammatory molecules, kinases, tubulins, amyloid-β aggre-
gates, adhesion molecules, growth factors, receptor proteins, protofilaments, prion
proteins, etc. The experimentally reported dipole moment [84] and log P values
(partition coefficient in octanol and water system) of curcumin (Table 1.1) indicate
that the molecule has partial charge transfer character and is moderately polar to be
soluble in lipid-like systems. Because of these properties and flexibility in its
structure, curcumin binds to most of the biomolecules. The hydrophobic interac-
tions and hydrogen-bonding interactions are mainly responsible for the efficient
binding. It is still premature to clarify the role of any single moiety for these
interactions but it appears that the orientation of the enolic group plays a crucial
role.
The α,β-unsaturated β-diketo moiety of curcumin participates in nucleophilic
addition reactions with molecules having functional groups like –SH, –SeH. This
1,4-addition reaction known as Michael addition reaction is of great significance in
curcumin biology, like it reacts with glutathione (GSH) and depletes the GSH in
cells [13]. Similarly through the reaction with the –SeH group, it inhibits thiore-
doxin reductase, an enzyme involved in cellular redox homeostasis [36].
Curcumin undergoes a fast chemical degradation in solution, where products like
ferulic aldehyde, ferulic acid, vanillin, etc. are formed [102, 116]. The degradation
occurs through the β-diketo moiety of curcumin and is increased in presence of light
and decreased when solubilised in aqueous solutions along with lipids, proteins,
surfactants, cyclodextrins, starch etc.
1 Curcumin and Its Role in Chronic Diseases 5
Curcumin binds to many metals and metalloproteins, and the binding is through
covalent interactions, as the diketo group is an excellent metal chelator [89, 90,
117]. Binding of curcumin to metals ions has several biological consequences. Its
binding to Al3+ ions is proposed to be one of the key factors involved in its role in
preventing the pathogenesis of Alzheimer’s disease (AD) [58]. Zn2+-curcumin
complex showed anticancer activity and Au3+ complexes exhibit anti-arthritic
activity [99]. Curcumin–metal chelation can be used to reduce toxicity of heavy
metals like Hg2+, Cd2+, Pb2+ [1, 81]. Several mixed ligand complexes of curcumin
with metals like Cu2+, Ni2+, Mn3+, Pd2+ are also finding applications as antioxi-
dants, superoxide dismutase mimics and anticancer agents [15, 114]. Recently
curcumin derived radiopharmaceuticals are being prepared and explored as new
diagnostic and therapeutic agents. For example, 99mTc4+ and 68Ga3+ complexes of
curcumin reported to bind to amyloid-β (Aβ) fibrils and plaques, are being explored
as novel radiodiagnostic agents for AD [12, 95].
The major issue concerning the development of curcumin based drugs is its
extremely low bioavailability [9, 86]. Due to relatively low intestinal absorption
and rapid metabolism in the liver, the oral bioavailability of curcumin is very low,
while most of it is excreted through the feces within 3–6 h after administration.
Even after oral administration in grams, no significant curcumin was detected in the
plasma, and the highest curcumin levels were found in the intestines and detectable
amounts were observed in the serum, but they fall below detection limit in other
tissues. However, i.p. and intravenous (i.v.) administration has shown better
6 A. Kunwar and K.I. Priyadarsini
Growth factors/cytokines are proteins which upon binding to their specific receptors
present on the plasma membrane of the cells stimulate multiple signal transduction
pathways leading to the expression of genes controlling various cellular processes
such as cell cycle, division, apoptosis, movement, inflammatory response to
external stimuli, etc. One of the hallmarks of a cancer cell is the constitutive or
increased expression of growth factors and their receptors inducing signals for the
uncontrolled proliferation or the growth [46]. Recent studies provide evidence that
curcumin targets such growth factors and their receptors to inhibit the growth and
proliferation of cancer cells [4, 6, 49, 110]. For example, curcumin was reported to
inhibit the effects of epidermal growth factor (EGF) and insulin growth factor
(IGF) by downregulating the expression and tyrosine kinase activity of EGF and
IGF receptors in colon cancer cells [93] and MCF-7 breast cancer cells [118].
Similarly curcumin has also been shown to downregulate the expression and
activity of HER-2/erbB2/neu/p185, another member of the EGF receptor super
family closely associated with breast, lung, kidney, and prostate cancers [52].
Further, the overexpression of other growth factors such as platelet-derived
growth factor (PDGF), vascular endothelial growth factor (VEGF) and their
receptors PDGFR and VEGFR, respectively have also been seen in many types of
solid tumors [28, 39]. Park et al. [82] showed that curcumin inhibits
PDGFR-induced proliferation of human hepatic myofibroblasts. In another study
curcumin was shown to block the angiogenesis or the formation of new blood
vessels in various cancer types by inhibiting both VEGF and VEGFR [28].
Some reports indicate that TNF acts as a growth factor for tumor cells.
Similarly other cytokines such as IL6 and IL2 regulate the proliferation of T cells
through the autocrine stimulation and play important roles in mounting the
8 A. Kunwar and K.I. Priyadarsini
Protein kinases are a class of enzymes, which activate the downstream signaling
molecules through phosphorylation using ATP as a source of inorganic phosphate
(Pi). They are the key transducers such as mitogen activated protein kinases
(MAPKs)/extracellular signal regulated kinases (ERKs), protein kinase A (PKA),
protein kinase B (PKB)/AKT, and protein kinase C (PKC), phosphatidyl inositol 3
phosphate kinase (PI3K), and mammalian target of rapamycin (mTOR) controlling
cell growth, proliferation, cytoprotection, cytokines production and death [3, 4, 49,
110]. Interestingly, curcumin has been shown to modulate the expression and
activity of MAPKs including ERKs, C-Jun, N terminal kinases (JNKs), p38
kinases, and the stress activated protein kinases (SAPK) in suppressing inflam-
mation and cancer [49]. Although inhibitory effect of curcumin on MAPKs has
been documented in several independent studies, there are also some reports, which
indicate that curcumin activates JNKs leading to apoptosis or cell death in tumor
cells [29]. Further, PI3K is the upstream regulator of PKB/AKT and the mTOR
pathways playing a central role in the genesis of cancer. Recent studies have shown
that curcumin inhibits proliferation/growth and induces apoptosis in tumor cells by
inhibiting mTOR pathway and it is mediated through the suppression of PI3K and
PKB/AKT. Additionally, the inhibitory effect of curcumin on mTOR signaling has
also been attributed to its ability to activate adenosine monophosphate kinase
(AMPK1), which senses cellular ATP levels and inhibits mTOR indirectly [122]. In
a separate study, curcumin was shown to block the signals transmitted from EGFR
by inhibiting the PI3K/AKT pathway in colon carcinoma cells [54]. On the con-
trary, curcumin activates the same PI3K and PKB/AKT signaling to induce the
expression of antioxidant genes in pre-cancer cells and thus prevents both tumor
initiation and promotion, a critical stage in carcinogenesis [61, 64]. Further, cur-
cumin also modulates PKC differentially, depending on the cell type and nature of
exogenous/endogenous stimuli. For example it has been shown to inhibit the tumor
promoting action of 12-o-tetradecanoylphorbol-13-acetate (TPA) by inactivating
PKC through oxidizing the vicinal thiols present within catalytic domain [72]. On
the other hand, the chemopreventive activity of curcumin has been linked with its
ability to activate PKC and the downstream p38 MAPK in human monocytes
leading to induction of antioxidant genes [94].
1 Curcumin and Its Role in Chronic Diseases 9
Transcription factors are the cellular proteins, which upon activation through sig-
naling cascades bind to the promoter/enhancer regions of their target genes and
regulate their transcription/expression. Curcumin by modulating various signal
transduction pathways affects the activation of a number of transcription factors
such as nuclear factor-κB (NF-κB), activator protein-1 (AP-1), early growth
response-1 (Egr-1), signal transducer and activator of transcription (STAT), per-
oxisome proliferator activated receptor-gamma (PPAR-γ), nuclear factor erythroid
2-related factor 2 (Nrf2), beta (β)-catenin and tumor suppressor p53 [19, 20, 24, 26,
57, 108, 110].
The NF-κB is reported to be constitutively activated in a variety of cancers and
inflammatory disorders. Researchers working on the various aspects of biological
activity have unanimously shown that curcumin is a strong suppressor of NF-κB
activation [104, 108]. The inactivation of NF-κB by curcumin is mediated through
the inhibition of IκBα kinase (IKK) directly and/or by modulating upstream sig-
naling cascade [108]. AP-1 is another transcription factor associated with growth
regulation, cell transformation, inflammation, and innate immune response.
Curcumin suppresses the activation of AP-1 either by inhibiting the upstream
kinases such as JNK, p38 and ERK or by directly interacting with AP-1 DNA
binding motifs present in the promoter regions of the target genes [20]. The tran-
scription factor Egr-1 is one of the immediate early induced gene products in
response to growth factor or carcinogen mediated signaling. Curcumin suppresses
the induction/de novo synthesis of Egr-1 protein in different cell types such as
endothelial cells, fibroblast, and colon cancer cells [26]. The family of STAT pro-
teins (STAT1-STAT7) performs the dual function of signal transducer as well as the
transcription factor. Of the seven STAT proteins, the elevated activity of STAT3 and
STAT5 has been mainly implicated in the angiogenic response and in growth of
various cancer types. Curcumin has been found to inhibit the phosphorylation of
STAT3 and STAT5 by upstream kinases (like janus kinase (JAK) and/or the growth
factor/G-protein-coupled receptor kinases) and their subsequent translocation to
nucleus there by suppressing the tumor cell proliferation and the pro-inflammatory
immune responses [19, 25]. The protein PPAR-γ also plays the dual role of nuclear
receptors of hormones and transcription factor. Curcumin has been shown to induce
the expression of PPAR-γ in various cell types such as colon cancer cells [24] and
hepatocytes leading to suppression of tumor cell proliferation, and the prevention of
liver fibrosis and sepsis [119]. The transcription factor Nrf2 induces the expression
of genes related to cytoprotection, antioxidant enzymes and phase II detoxification
enzymes. Curcumin induces the antioxidant and cytoprotective responses in various
cell types such as epithelial, monocytes, hepatocytes by activating Nrf2. The
mechanism of activation is either through directly interacting with Keap-1 (inhibitor
of Nrf2) or by positively regulating upstream kinases such as PI3K, PKC and MAPK
[53, 94]. The β-catenin and p53 are some of other important transcription factors,
which control the expression of genes involved in cell cycle progression and check
10 A. Kunwar and K.I. Priyadarsini
points. Curcumin has been reported to induce as well as inhibit the transcriptional
activity of β-catenin by differentially modulating the activity of glycogen synthase
kinase-3 (GSK3) [57, 123]. The inhibition of β-catenin activation by curcumin has
been liked with the suppression of tumor cell proliferation and the induction of
apoptosis. Whereas curcumin mediated activation of β-catenin has been attributed
toits anti-AD activity. The p53 protein is a tumor suppressor protein. Depending on
cell type, curcumin regulates the level of p53 in different ways. For example, in
normal thymocytes and myeloid leukemic cells, curcumin downregulates the
expression of p53 and inhibits p53-induced apoptosis [45, 112]. In contrast to such
studies, curcumin has been shown to upregulate p53 in colon, neuroblastoma,
lymphoma, prostate, and human breast cancer cells to induce apoptosis [27, 110].
It has been confirmed by several researchers that oxidative stress and oxidative
damage are directly associated with chronic inflammation, which in turn leads to
several chronic diseases, like cancer, metabolic, neurological, pulmonary, intestinal
and cardiovascular diseases. Since curcumin has been identified as an important
remedy against oxidative stress and inflammation, extensive research work has been
undertaken to use curcumin against many chronic diseases. Important examples are
mentioned briefly here [3, 51, 101].
Inflammatory bowel disease (IBD), symptomatic with severe diarrhea, pain,
fatigue and weight loss, involves chronic inflammation of all or part of digestive
tract. Both turmeric extract and polymer loaded curcumin formulations have been
examined in animals and humans for treatment against IBD and many encouraging
results were obtained without any side effects [18, 47, 48, 111]. In mice, oral
administration of curcumin (100 mg/kg per day) for ten consecutive days prior to
the induction of colitis by acetic acid and further continuation for 2 days showed
decreased colon injury and ameliorated macroscopic and microscopic colitis sores.
IBD patients treated with curcumin (550 mg twice daily for 1 month) showed
decreased symptoms and inflammation indices, without any significant side effects.
Rheumatoid arthritis (RA), a long-lasting autoimmune disorder that primarily
affects joints on both sides of the body, is a systemic chronic inflammatory disorder.
To reduce arthritic reaction, it is essential to start early treatment and combination
therapy. As a nonsteroid anti-inflammatory drug and antioxidant, curcumin has
been examined as a potential therapy for RA, accordingly RA patients treated with
curcumin showed significant symptomatic improvement [76, 90]. Similarly cur-
cumin (500 mg) alone and in combination with diclofenac sodium (50 mg)
administered in patients with RA showed best improvement in the overall disease
scores. In a short term, a double-blind crossover study involving 18 young patients
suffering from RA, oral curcumin at a daily dose of 12 g for 2 weeks exerted an
anti-RA activity comparable to that of a standard drug phenylbutazone.
Diabetes mellitus (DM), commonly referred to as diabetes, is a metabolic disease
in which there are high blood sugar levels over a prolonged period. Serious
long-term complications include cardiovascular disease, stroke, chronic kidney
failure, foot ulcers, and damage to the eyes. Effect of curcumin against diabetes was
successfully demonstrated first in one patient in 1972 [109] and since then more
than 200 papers have been published in this related subject [2, 73, 124]. Curcumin
1 Curcumin and Its Role in Chronic Diseases 13
has been shown to reduce blood glucose and glycosylated hemoglobin levels and
prevented weight loss in rodent models. Oral administration of curcumin
(80 mg/kg) showed anti-hyperglycemic effect and improved insulin sensitivity. It
was also effective in improving glucose intolerance. One or two contradicting
effects were also reported, where intragastric administration of curcumin did not
show any effect in blood glucose. Curcumin has been shown to reduce several other
complications associated with diabetes like fatty liver, diabetic neuropathy, diabetic
nephropathy, vascular diseases, musculoskeletal diseases and also islet viability.
Neurodegenerative diseases including Parkinson’s and AD occur due to the
progressive loss of structure or function of neurons, including death of neurons.
Curcumin was considered as a potent drug mainly due to the fact that the epi-
demiological studies indicating reduced risk of AD amongst Indians consuming
turmeric in their diet. In line with this, several experimental studies in mice models
confirmed anti-AD effects of curcumin [44]. It significantly lowered the levels of
oxidized proteins, IL-1β, insoluble and soluble plaque burden in AD transgenic
Tg2576 mice brain. There are several ongoing clinical trials on the effect of cur-
cumin for the prevention of AD. In one such study, daily intake of curcumin 1–
4 g/day over 6 months found a trend towards increased serum Aβ levels which is
considered as a possible clearance of Aβ plaques from the brain, but improvement
in cognitive performance was not observed during the 6 months trial [16]. A few
other studies indicate that curcumin also exhibits antidepressant and neuroprotec-
tive properties [32, 69, 75].
Psoriasis, a chronic skin disease, occurs when the immune system overreacts,
causing inflammation and flaking of skin and is characterized by thick red and scaly
lesions on any part of the body. Since ancient times turmeric has been used as a skin
protectant in the Indian subcontinent. Further experimental evidences strongly
indicate that curcumin is a potential natural product to suppress psoriasis by
inhibiting the keratinocyte proliferation [59]. In a clinical trial, orally administered
curcumin demonstrated therapeutic effects with no adverse reaction in patients with
psoriasis and reduced psoriasis area and severity index [66]. Curcumin skin for-
mulations are being marketed for skin diseases including psoriasis.
In addition to these important diseases, curcumin has been examined for treat-
ment of other chronic diseases like cardiovascular diseases, allergy, asthma,
bronchitis, kidney diseases, obesity, scleroderma, vitiligo, peptic ulcer, pylori
infection and ophthalmic disorders and encouraging results are reported [3, 51]. The
anticancer effects are discussed in detail in the next section.
drugs
Ongoing 22 USA, Puerto Rico, Canada, UK,
Korea, India, Iran, Israel, Italy,
France, Austria, Belgium
Withdrawn 4 USA
Inflammatory bowel disease (IBD)/H. pylori infection/hepatic injury Completed 5 USA, Israel, Korea Reduces the levels of inflammatory
cytokines and symptoms
Ongoing 6 Israel, France, Itlay
Withdrawn
Cardiovascular diseases and metabolic syndrome Completed 7 France, Iran, Germany, Canada Decreases serum levels of cholesterol and
lipid peroxides, low density lipoprotein
(LDL), ApoB
Increases high density lipoprotein (HDL),
ApoA
Ongoing 5 Thailand, Canada, USA, Singapore
15
Withdrawn 1 USA
(continued)
Table 1.3 (continued)
16
Ongoing
Withdrawn
Psoriasis Completed 2 USA Decreases phosphorylase kinase activity a
marker of psoriasis disease
Ongoing 1 Italy
Withdrawn
Bioavailability and pharmacokinetics Completed 10 Germany, USA, Austria, Canada, Safe even at 12 g/day
Italy Poor bioavailability in serum and tissues
Ongoing 2 USA, India
Withdrawn 2 Israel, USA
General health benefits Completed 8 USA, France, Korea Dietary supplementation improves the levels
of antioxidant enzymes and reduces
oxidative stress
Improves general health
17
gemcitabine resistant, patients were safe and seem to tolerate well [34, 60]. In a
open-label phase I clinical trial, 14 patients with advanced and metastatic breast
cancer were examined for feasibility and tolerability of curcumin in combination
with docetaxel, based on which it has been recommended that 6 g curcumin/day for
seven consecutive days for 3 weeks would be safe in combination with a standard
dose of docataxel [17].
In a randomized double-blind study, curcumin (100 mg) in combination with
isoflavin (40 mg) for 6 months in 85 patients showed synergism and suppressed
prostrate specific antigen after treatment [55]. Curcumin was also examined against
multiple myeloma and two clinical trials were completed. In one single blinded
crossover pilot study in 26 patients with monoclonal gammopathy of undetermined
significance (MGUS), curcumin showed therapeutic potential against MGUS [41].
In another study, involving 29 patients with asymptomatic, relapsed multiple
myeloma, curcumin was found to be effective either alone or in combination with
bioperine [113]. In a randomized open-label study involving 25 patients with
chronic myeloid leukemia, curcumin (5 g) administered with imatinib (0.8 g)
showed better efficacy in decreasing the nitric oxide levels. This suggested that
curcumin can be used as an adjuvant to imatinib for the treatment of chronic
myeloid leukemia patients [40]. Curcumin has also showed encouraging results in
combination with other cancer drugs like paclitaxel, and cisplatin.
Oral curcumin has also been shown to prevent the H. pylori infection, a pre-
cursor of gastric cancer, prostatic intraepithelial neoplasia, a precursor of prostate
cancer and to reduce the poly numbers and size in patients with FAP. The efficacy
of curcumin against IBD has already been established in clinical trials. Similarly
curcumin could effectively induce the gallbladder emptying and reduce gall stone
formation, a potential risk factor for gallbladder cancer. In another study, oral
curcumin (400 mg three times a day) compared to placebo showed much better
anti-inflammatory response against spermatic cord edema in patients that underwent
surgical repair of hernia and/or hydrocele.
Many other clinical trials against variety of other diseases have also been ini-
tiated and are being continued, interested readers may refer to some of the recent
reviews on curcumin clinical trials [4, 43, 97, 121].
1.7 Conclusions
Curcumin
Liposomal curcumin
Curcumin mouthwash
and well tolerated by the patients under study. Its efficacy against diabetes,
inflammatory bowel disease and arthritis are well proven in the clinic. Extensive
clinical trials to develop curcumin as anticancer drug are still continuing, and the
results are moderately encouraging, mainly due to complexity of the disease and
low bioavailability of curcumin. To partly overcome this, new curcumin formula-
tions with improved bioavailability are successfully designed and developed. More
clinical trials with larger participants are warranted to translate the preclinical
research to the clinical cancer medicine. Curcumin- and turmeric-based nutraceu-
tical formulations are becoming very popular and more than hundred varieties of
products in the form of drinks, capsules, creams, gels, food supplements are
available in the market (Fig. 1.3 gives a few representative products). These
products are recommended for both healthy people and also for patients undergoing
treatment of chronic diseases. In view of all these gifted properties; it is not
exaggerating to say that this inexpensive and innocuous dietary agent, a highly
pleiotropic molecule, will be a “Panacea” in near future.
Acknowledgments The authors wish to express sincere thanks to Department of Atomic Energy,
Government of India and acknowledge the contributions of many co-authors and students whose
names appeared in the publications listed from our group.
References
1. Agarwal R, Goel SK, Behari JR (2010) Detoxification and antioxidant effects of curcumin in
rats experimentally exposed to mercury. J Appl Toxicol 30:457–468
2. Aggarwal BB (2010) Targeting inflammation induced obesity and metabolic diseases by
curcumin and other nutraceuticals. Annu Rev Nutr 30:173–199
20 A. Kunwar and K.I. Priyadarsini
3. Aggarwal BB, Sung B (2009) Pharmacological basis for the role of curcumin in chronic
diseases: an age-old spice with modern targets. Trends Pharmacol Sci 30:85–94
4. Aggarwal BB, Kumar A, Bharti AC (2003) Anticancer potential of curcumin: preclinical and
clinical studies. Anticancer Res 23:363–398
5. Aggarwal BB, Shishodia S, Takada Y et al (2005) Curcumin suppresses the paclitaxel
induced nuclear factor-κB pathway in breast cancer cells and inhibits lung metastasis of
human breast cancer in nude mice. Clin Cancer Res 11:7490–7498
6. Aggarwal BB, Sundaram C, Malini N et al (2007) Curcumin: the Indian solid gold. Adv Exp
Med Biol 595:243–305
7. Aggarwal BB, Gupta SC, Sung B (2013) Curcumin: an orally bioavailable blocker of TNF
and other pro-inflammatory biomarkers. Br J Pharmacol 169(8):1672–1692
8. Alrawaiq NS, Abdullah A (2014) A review of antioxidant polyphenol curcumin and its role
in detoxification. Int J PharmTech Res 6:280–289
9. Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB (2007) Bioavaialbility of
curcumin: problems and promises. Mol Pharm 4:807–818
10. Arbiser JL, Klauber N, Rohan R, van LR, Huang MT, Fisher C, Flynn E, Byers HR (1998)
Curcumin is an in vivo inhibitor of angiogenesis. Mol Med 4:376–383
11. Asai A, Miyazawa T (2000) Occurrence of orally administered curcuminoid as glucuronide
and glucuronide/sulfate conjugates in rat plasma. Life Sci 67:2785–2793
12. Asti M, Ferrari E, Groci S et al (2014) 68Ga-labelled curcuminoids complexes:
characterisation of potential radiotracers for imaging of Alzheimer’s disease. Inorg Chem
53:4922–4933
13. Awasthi A, Pandya U, Singhal SS et al (2000) Curcumin–glutathione interactions and the
role of human glutathione S-transferase PI-1. Chem Biol Interact 128:19–38
14. Bae MK, Kim SH, Jeong JW et al (2006) Curcumin inhibits hypoxia-induced angiogenesis
via down-regulation of HIF-1. Oncol Rep 15:1557–1562
15. Barik A, Mishra B, Shen L et al (2005) Evaluation of new copper–curcumin complex as
superoxide dismutase mimic and its free radical reactions. Free Radic Biol Med 39:811–822
16. Baum L, Lam CW, Cheung SK et al (2008) Six month randomized placebo controlled double
blind pilot clinical trial of curcumin in patients of Alzheimer disease. J Clin
Psychopharmacol 28:110–113
17. Bayet-Robert M, Kwaitkowski F, Leheurteur M et al (2010) Phase I dose escalation trial of
docataxel plus curcumin in patients with advanced metastatic breast cancer. Cancer Biol Ther
9:8–14
18. Beloqui A, Coco R, Memvanga PB et al (2014) pH sensitive nanoparticles for colonic
delivery in inflammatory bowel disease. Int J Pharm 473:203–212
19. Bharti AC, Donato N, Aggarwal BB (2003) Curcumin (diferuloylmethane) inhibits
constitutive and IL-6-inducible STAT3 phosphorylation in human multiple myeloma cells.
J Immunol 171:3863–3871
20. Bierhaus A, Zhang Y, Quehenberger P et al (1997) The dietary pigment curcumin reduces
endothelial tissue factor gene expression by inhibiting binding of AP-1 to the DNA and
activation of NF-κB. Thromb Haemost 77:772–782
21. Chan MM, Huang HI, Fenton MR, Fong D (1998) In vivo inhibition of nitric oxide synthase
gene expression by curcumin: a cancer preventive natural product with anti-inflammatory
properties. Biochem Pharmacol 55:1955–1962
22. Chattopadhyay I, Biswas K, Bandyopadhyay U et al (2004) Turmeric and curcumin:
biological actions and medicinal applications. Curr Sci 87:44–53
23. Chattopadhyay I, Bandyopadhyay U, Biswas K et al (2006) Indomethacin inactivates gastric
peroxidase to induce reactive-oxygen-mediated gastric mucosal injury and curcumin protects
it by preventing peroxidase inactivation and scavenging reactive oxygen. Free Radic Biol
Med 40:1397–1408
24. Chen A, Xu J (2005) Activation of PPARγ by curcumin inhibits Moser cell growth and
mediates suppression of gene expression of cyclin D1 and EGFR. Am J Physiol Gastrointest
Liver Physiol 288:G447–G456
1 Curcumin and Its Role in Chronic Diseases 21
25. Chen WH, Chen Y, Cui GH et al (2004) Effect of curcumin on STAT5 signaling pathway in
primary CML cells. Zhonghua Xue Ye Xue Za Zhi 12:572–576
26. Chen A, Xu J, Johnson AC (2006) Curcumin inhibits human colon cancer cell growth by
suppressing gene expression of epidermal growth factor receptor through reducing the
activity of the transcription factor Egr-1. Oncogene 25:278–287
27. Choudhuri T, Pal S, Das T, Sa G (2005) Curcumin selectively induces apoptosis in
deregulated cyclin D1-expressed cells at G2 phase of cell cycle in a p53-dependent manner.
J Biol Chem 280:20059–20068
28. Chua CC, Hamdy RC, Chua BH (2000) Mechanism of transforming growth
factor-β1-induced expression of vascular endothelial growth factor in murine osteoblastic
MC3T3-E1 cells. Biochim Biophys Acta 1497:69–76
29. Collett GP, Campbell FC (2004) Curcumin induces c-Jun N-terminal kinase-dependent
apoptosis in HCT116 human colon cancer cells. Carcinogenesis 25:2183–2189
30. Conney AH (2003) Enzyme induction and dietary chemicals as approaches to cancer
chemoprevention: the Seventh DeWitt S. Goodman lecture. Cancer Res 63:7005–7031
31. Dhillon N, Aggarwal BB, Newman RA et al (2008) Phase II trial of curcumin in patients with
advanced pancreatic cancer. Clin Cancer Res 14:4491–4499
32. Kim DS, Kim JY, Han Y (2012) Curcuminoids in neurodegenerative diseases. CNS drug
Discov 7:184–204
33. Durgaprasad S, Pai CG, Kumar V et al (2005) A pilot study of the antioxidant effect of
curcumin in tropical pancreatic. Ind J Med Res 122:315–318
34. Epelbaum R, Schaffer M, Vizel B et al (2010) Curcumin and gemcitabine in patients with
advanced pancreatic cancer. Nutr Cancer 62:1137–1141
35. Esatbeyoglu T, Huebbe P, Insa MA et al (2012) Curcumin—from molecule to biological
function. Angew Chem Int Ed 51:5308–5332
36. Fang J, Jun L, Holmegren A (2005) Thioredoxin reductase is irreversibly modified by
curcumin: a novel molecular mechanism for its anticancer activity. J Biol Chem 280:25284–
25290
37. Garcea G, Jones DJ, Singh R et al (2004) Detection of curcumin and its metabolites in
hepatic tissue and portal blood of patients following oral administration. Br J Cancer
90:1011–1015
38. Garcea G, Berry DP, Jones DJ et al (2005) Consumption of the putative chemopreventive
agent curcumin by cancer patients: assessment of curcumin levels in the colorectum and their
pharmacokinetic consequences. Cancer Epidemiol Biomarkers Prev 14:120–125
39. George D (2003) Targeting PDGF receptors in cancer–rationales and proof of concept
clinical trials. Adv Exp Med Biol 532:141–151
40. Ghalaut VS, Sangwan L, Dahiya K et al (2012) Effect of imatinib therapy with and without
turmeric powder on nitric oxide levels in chronic myeloid leukemia. J Oncol Pharm Pract
18:186–190
41. Golombick T, Diamond TH, Badmaev V et al (2009) The potential role of curcumin in
patients with monoclonal gammopathy of undefined significance—its effect on
paraproteinemia type I collagen bone turnover marker. Clin Cancer Res 15:5917–5922
42. Gupta S, Prasad S, Ji HK et al (2011) Multitargeting by curcumin as revealed by molecular
interaction studies. Nat Prod Rep 28:1937–1955
43. Gupta SC, Patchva S, Aggarwal BB (2013) Therapeutic potential of curcumin: lessons
learned from clinical trials. AAPS J 15:195–218
44. Hamaguchi T, Ono K, Yamada M (2010) Review: curcumin and Alzheimer’s disease. CNS
Neurosci Ther 16:285–297
45. Han SS, Chung ST, Robertson DA, Ranjan D, Bondada S (1999) Curcumin causes the
growth arrest and apoptosis of B cell lymphoma by downregulation of egr-1, c-myc, bcl-XL,
NF-κB, and p53. Clin Immunol 93:152–161
46. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:
646–674
22 A. Kunwar and K.I. Priyadarsini
47. Hanai H, Sugimoto K (2009) Curcumin has prospects for the treatment of inflammatory
bowel disease. Curr Pharma Des 15:2087–2094
48. Hanai H, Iida T, Takeuchi K et al (2006) Curcumin maintenance therapy for ulcerative colitis
randomized multicenter, double-blind placebo controlled trial. Clin Gastroentero Heatol
4:1502–1506
49. Hasima N, Aggarwal BB (2012) Cancer-linked targets modulated by curcumin. Int J
Biochem Mol Biol 3:328–351
50. He ZY, Shi CB, Wen H et al (2011) Upregulation of p53 expression in cancer patients with
colorectal cancer by administration of curcumin. Cancer Investig 29:208–213
51. He ZY, Yue Y, Zheng X, Zhang K, Chen S, Du Z et al (2015) Curcumin, inflammation, and
chronic diseases: how are they linked? Molecules 20(5):9183–9213
52. Hong RL, Spohn WH, Hung MC (1999) Curcumin inhibits tyrosine kinase activity of
p185neu and also depletes p185neu. Clin Cancer Res 5:1884–1891
53. Hoque M, Gong P, Killeen E, Green CJ, Foresti R, Alam J, Motterlini R (2003) Curcumin
activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive
element. Biochem J 371:887–895
54. Hussain AR, Al-Rasheed M, Manogaran PS et al (2006) Curcumin induces apoptosis via
inhibition of PI3′-kinase/AKT pathway in acute T cell leukemias. Apoptosis 11:245–254
55. Ide H, Tokiwa S, Sakamaki K et al (2010) Combined inhibitory effects of soy isoflavons and
curcumin on the production of prostate specific antigen. Prostate 70:1127–1133
56. Ireson RC, Jones DJL, Orr S et al (2002) Metabolism of the cancer chemopreventive agent
curcumin in human and rat intestine. Cancer Epidemol Biomarkers Prev 11:105–111
57. Jaiswal AS, Marlow BP, Gupta N et al (2002) Beta-catenin-mediated transactivation and
cell–cell adhesion pathways are important in curcumin (diferuylmethane)-induced growth
arrest and apoptosis in colon cancer cells. Oncogene 21:8414–8427
58. Jiang T, Zhi X, Zhang Y, Pan L, Zhou P (2012) Inhibitory effect of curcumin on the Al(III)-
induced A β(42) aggregation and neurotoxicity in vitro. Biochim Biophys Acta Mol Basis
Dis 1822:1207–1215
59. Jun S, Yi Z, Jinhong H (2013) Curcumin inhibits imiquimod induced psoriasis like
inflammation by inhibiting IL-1β and IL-6 production in mice. PLoS One 8:e67078
60. Kanai M, Yoshimura K, Asada M et al (2011) A phase I/II study of gemcitabine-based
chemotherapy plus curcumin for patients with gemcitabine-resistant pancreatic cancer.
Cancer Chemother Pharmacol 68:157–164
61. Kang ES, Woo IS, Kim HJ et al (2007) Up-regulation of aldose reductase expression
mediated by phosphatidylinositol 3-kinase/Akt and Nrf2 is involved in the protective effect
of curcumin against oxidative damage. Free Radic Biol Med 43:535–545
62. Kang ES, Kim GH, Kim HJ et al (2008) Nrf2 regulates curcumin-induced aldose reductase
expression indirectly via nuclear factor-κB. Pharmacol Res 58:15–21
63. Kawamori T, Lubet R, Steele VE et al (1999) Chemopreventive effect of curcumin, a
naturally occurring anti-inflammatory agent, during the promotion/progression stages of
colon cancer. Cancer Res 59:597–601
64. Khan N, Afaq F, Mukhtar H (2008) Cancer chemoprevention through dietary antioxidants:
progress and promise. Antioxid Redox Signal 10:475–510
65. Kunwar A, Barik A, Pandey R, Priyadarsini KI (2006) Transport of liposomal and albumin
loaded curcumin to living cells: an absorption and fluorescence spectroscopic study. Biochim
Biophys Acta (General) 1760:1513–1520
66. Kurd SK, Smith N, Van Voorhees A et al (2008) Oral curcumin in the treatment of moderate
to severe psoriasis vulgaris, a prospective clinical trial. J Am Acd Dermatol 58:625–631
67. Kuttan R, Sreedharan PC, Joseph CD (1987) Turmeric and curcumin as topical agents in
cancer therapy. Tumori 73:29–31
68. Lampe V, Milobedzka J (1913) Studien uber curucmin. Ber Dtsch Chem Ges 46:2235–2240
69. Lee WH, Loo CY, Bebawy M, Luk F, Mason RS, Rohanizadeh R (2013) Curcumin and its
derivatives: their application in neuropharmacology and neuroscience in the 21st century.
Curr Neuropharmacol 11(4):338–378
1 Curcumin and Its Role in Chronic Diseases 23
70. Li N, Chen X, Han C, Chen J (2002) Chemopreventive effect of tea and curcumin on
DMBA-induced oral carcinogenesis in hamsters. Wei Sheng Yan Jiu 31:354–357
71. Li L, Braiteh FS, Kurzrock R (2005) Liposome-encapsulated curcumin: in vitro and in vivo
effects on proliferation, apoptosis, signaling, and angiogenesis. Cancer 104:1322–1331
72. Liu JY, Lin SJ, Lin JK (1993) Inhibitory effects of curcumin on protein kinase C activity
induced by 12-O-tetradecanoyl-phorbol-13-acetate in NIH 3T3 cells. Carcinogenesis
14:857–861
73. Meng BI, Li J, Cao H (2013) Antioixidant and anti-inflammatory activities of curcumin on
diabetes mellitus and its complications. Curr Pharm Des 19:2101–2103
74. Milobedzka J, Kostanecki S, Lampe V (1910) Zur Kenntnis des Curcumins. Berichte der
Deutschen Chemischen Gessellschaft 43:2163–2170
75. Monroy A, Lithgow GJ, Alavez S (2013) Curcumin and neurodegenerative diseases.
BioFactors 39:122–132
76. Moon DO, Kim MO, Choi YH et al (2010) Curcumin attenuates inflammatory response in
IL-1 bets induced human synovial fibroblasts and collagen induced arthritis in mouse model.
Int Immunopharmacol 10:605–610
77. Mori Y, Tatematsu K, Koide A et al (2006) Modification by curcumin of mutagenic
activation of carcinogenic N-nitrosamines by extrahepatic cytochromes P-450 2B1 and 2E1
in rats. Cancer Sci 97:896–904
78. Noorafshan A, Ashkani-Esfahani S (2013) A review of therapeutic effects of curcumin. Curr
Pharm Des 19:2032–2046
79. Oppenheimer A (1937) Turmeric in biliary diseases. Lancet 229:619–621
80. Pabon HJ (1964) Synthesis of curcumin and related compounds. Rev Trav Chim 83:379–386
81. Pallikkavil R, Ummathur MS, Sreedharan S, Krishnankutty K (2013) Synthesis,
characterization and antimicrobial studies of Cd(II), Hg(II), Pb(II), Sn(II) and Ca(II)
complexes of curcumin. Main Group Met Chem 36:123–127
82. Park SD, Jung JH, Lee HW et al (2005) Zedoariae rhizome and curcumin inhibits
platelet-derived growth factor-induced proliferation of human hepatic myofibroblasts. Int
Immunopharmacol 5:555–569
83. Park W, Ruhul Amin ARM, Chen ZG, Shin DM (2013) New perspectives of curcumin in
cancer prevention. Cancer Prev Res 6:387–400
84. Párkányi C, Stem-Beren MR, Martı́nez OR, Aaron JJ, MacNair MB, Arrieta AF (2004)
Solvatochromic correlations and ground- and excited-state dipole moments of curcuminoid
dyes. Spectrochim Acta A 60:1805–1810
85. Perkins S, Verschoyle RD, Hill K et al (2002) Chemopreventive efficacy and
pharmacokinetics of curcumin in mouse, a model of familial adenomatous polyposis.
Cancer Epidemiol Biomarkers Prev 11:535–540
86. Prasad S, Tyagi AK, Aggarwal BB (2014) Recent developments in delivery, bioavailability,
absorption and metabolism of curcumin: the golden pigment from golden spice. Cancer Res
Treat 46:2–18
87. Priyadarsini KI (1997) Free radical reactions of curcumin in model membranes. Free Radic
Biol Med 23:838–884
88. Priyadarsini KI (2009) Photophysics, photochemistry and photobiology of curcumin: studies
from organic solutions, bio-mimetics and living cells. J Photochem Photobiol C Chem Rev
10:81–96
89. Priyadarsini KI (2013) Chemical and structural features influencing the biological activity of
curcumin. Curr Pharm Des 19:2093–2100
90. Priyadarsini KI (2014) The chemistry of curcumin: from extraction to therapeutic agent.
Molecules 19:20091–20112
91. Ramadan G, El-Menshawy O (2013) Protective effects of ginger–turmeric rhizomes mixture
on joint inflammation, atherogenesis, kidney disfunction and other complications in a rat
model of human rheumatoid arthritis. Int J Rheum Dis 16:219–229
24 A. Kunwar and K.I. Priyadarsini
92. Ranjan D, Chen C, Johnston H, Jeon H, Nagabhushan M (2004) Curcumin inhibits mitogen
stimulated lymphocyte proliferation, NFkappaB activation, and IL-2 signaling. J Surg Res
121:171–177
93. Reddy S, Rishi AK, Xu H et al (2006) Mechanisms of curcumin- and EGF-receptor related
protein (ERRP)-dependent growth inhibition of colon cancer cells. Nutr Cancer 55:185–194
94. Rushworth SA, Ogborne RM, Charalambos CA, O’Connell MA (2006) Role of protein
kinase C δ in curcumin-induced antioxidant response element-mediated gene expression in
human monocytes. Biochem Biophys Res Commun 341(4):1007–1016
95. Sagnou M, Benaki D, Triantis C et al (2011) Curcumin as the OO bidentate ligand in “2 + 1”
complexes with the [M(CO)(3)](+) (M = Re, 99mTc) tricarbonyl core for radiodiagnostic
applications. Inorg Chem 50:1295–1303
96. Schraufstatter E, Bernt H (1949) Antibacterial action of curcumin and related compounds.
Nature 164:456–457
97. Shanmugam MK, Rane G, Kanchi MM et al (2015) The multifaceted role of curcumin in
cancer prevention and treatment. Molecules 20:2728–2769
98. Sharma OP (1976) Antioxidant activity of curcumin and related compounds. Biochem
Pharmacol 25:1811–1812
99. Sharma KK, Chandra S, Basu DK (1987) Synthesis and antiarthritic study of a new orally
active diferuloyl methane (curcumin) gold complex. Inorg Chim Acta 135:47–48
100. Sharma RA, Euden SA, Platton SL et al (2004) Phase I clinical trial of oral curcumin:
biomarkers of systemic activity and compliance. Clin Cancer Res 10:6847–6854
101. Shehzad A, Rehman G, Lee YS (2013) Curcumin in inflammatory diseases. BioFactors
39:69–77
102. Shen L, Ji HF (2012) The pharmacology of curcumin: is it the degradation products? Trends
Mol Med 18:138–143
103. Shen G, Xu C, Hu R et al (2006) Modulation of nuclear factor E2-related factor 2—mediated
gene expression in mice liver and small intestine by cancer chemopreventive agent curcumin.
Mol Cancer Ther 5:39–51
104. Shishodia S, Amin HM, Lai R, Aggarwal BB (2005) Curcumin (diferuloylmethane) inhibits
constitutive NF-κB activation, induces G1/S arrest, suppresses proliferation, and induces
apoptosis in mantle cell lymphoma. Biochem Pharmacol 70:700–713
105. Shoba G, Joy D, Joseph T, Majeed M, Rajendran R, Srinivas PSSR (1998) Influence of
piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med
64:353–356
106. Shrimal RC, Dhawan BN (1973) Pharmacology of diferuloyl methane (curcumin), a
non-steroidal anti-inflammatory agent. J Pharm Pharmacol 25:447–452
107. Singh S (2007) From exotic spice to modern drug? Cell 130:765–768
108. Singh S, Aggarwal B (1995) Activation of transcription factor NF-κB is suppressed by
curcumin (Diferuloylmethane). J Biol Chem 270:24995–25000
109. Srinivasan M (1972) Effect of curcumin on blood sugar as seen in a diabetic subject. Ind J
Med Sci 26:269–270
110. Strimpakos AS, Sharma RA (2008) Curcumin: preventive and therapeutic properties in
laboratory studies and clinical trials. Antioxid Redox Signal 10:511–545
111. Taylor RA, Leonard MC (2011) Curcumin for inflammatory bowel disease, a review of
human studies. Alt Med Rev 16:152–156
112. Tsvetkov P, Asher G, Reiss V, Shaul Y, Sachs L, Lotem J (2005) Inhibition of NAD(P)H:
quinone oxidoreductase 1 activity and induction of p53 degradation by the natural phenolic
compound curcumin. Proc Natl Acad Sci USA 102:5535–5540
113. Vadhan-Raj S, Weber D, Wang M et al (2007) Curcumin downregulates NF-κB and related
genes in patients with multiple myeloma: results of phase 1/2 study. Blood 110:357a
114. Vajragupta O, Boonchoong P, Watanabe H et al (2003) Manganese complexes of curcumin
and its derivatives: evaluation for the radical scavenging ability and neuroprotective activity.
Free Radic Biol Med 35:1632–1644
115. Vogel HA, Pelletier J (1815) Curcumin-biological and medicinal properties. J Pharma 2:50
1 Curcumin and Its Role in Chronic Diseases 25
116. Wang YJ, Pan MH, Cheng AL et al (1997) Stability of curcumin in buffer solution and
characterization of its degradation products. J Pharma Biomed Anal 15:1867–1876
117. Wanninger S, Lorenz V, Subhan A, Edelmann FT (2015) Metal complexes of curcumin—
synthetic strategies, structures and medicinal applications. Chem Soc Rev 44:4986–5002
118. Xia Y, Jin L, Zhang B et al (2007) The potentiation of curcumin on insulin-like growth
factor-1 action in MCF-7 human breast carcinoma cells. Life Sci 80:2161–2169
119. Xu J, Fu Y, Chen A (2003) Activation of peroxisome proliferator-activated receptor gamma
contributes to the inhibitory effects of curcumin on rat hepatic stellate cell growth. Am J
Physiol Gastrointest Liver Physiol 285:G20–G30
120. Yallapu MM, Ebeling MC, Khan S et al (2013) Novel curcumin-loaded magnetic
nanoparticles for pancreatic cancer treatment. Mol Cancer Ther 12:1471–1480
121. Yang C, Su X, Liu A et al (2013) Advances in clinical study of curcumin. Curr Pharm Des
19:1966–1973
122. Yu S, Shen G, Kong TA (2006) Curcumin inhibits mTOR signaling by inhibiting protein
kinase B/Akt and activating AMP-activated protein kinase (AMPK) in prostate cancer cell
line PC-3. Proc Am Assoc Cancer Res 47:538
123. Zhang X, Yin WK, Shi XD, Li Y (2011) Curcumin activates Wnt/β-catenin signaling
pathway through inhibiting the activity of GSK-3β in APPswe transfected SY5Y cells. Eur J
Pharm Sci 42:540–546
124. Zhang D-W, Fu M, Gao S-H, Liu J-L (2013) Curcumin and diabetes: a systematic review.
Evid Complement Altern Med 16:1–22
Chapter 2
Berberine and Its Role in Chronic Disease
2.1 Introduction
Cardiovascular diseases are yet the most common causes of death and one of the
first causes of disability in industrialized countries and despite the efforts towards
primary prevention of cardiovascular disease, many patients still remain at risk [1].
Lifestyle interventions such as diet and/or physical activity are the most
barberry was used to treat gall bladder and liver problems, while it was used in the
treatment of abnormal uterine bleeds and rheumatism in Russia and Bulgaria [18,
19]. In North America, the Eclectics used barberry for treatment of malaria and as a
general tonic [20]. Also, the American Indians found it effective in improving
appetite and used its dried fruit as a gargle [21, 22].
Medicinal properties for all parts of the plant have been reported, including tonic,
antimicrobial, antiemetic, antipyretic, antipruritic, antioxidant, anti-inflammatory,
hypotensive, antiarrhythmic, sedative, antinociceptive, anticholinergic and chola-
gogue actions, and it has been used in some cases like cholecystitis, cholelithiasis,
jaundice, dysentery, leishmaniasis, malaria and gall stones [23]. Furthermore, ber-
berine has been used for treating diarrhoea and gastrointestinal disorders for a long
time [24, 25]. It has multiple pharmacological effects including; antimicrobial activity
against 54 microorganisms [26], inhibition of intestinal ion secretion and smooth
muscle contraction, inhibition of ventricular tachyarrhythmia, reduction of inflam-
mation, stimulation of bile secretion and bilirubin discharge [27].
Berberine has low bioavailability and poor absorption through the gut wall
(<5 %) and bowel P-glycoprotein contributes to that, actively expelling the alkaloid
from the lumen mucosal cells [28].
In a rat noncompartmental model [29], unbound berberine is transported to bile
through active transportation and it is metabolized by P450 enzyme system in liver,
with phase I demethylation and phase II glucuronidation. Berberine has four main
metabolites identified in rats: berberrubine, thalifendine, demethyleneberberine and
jatrorrhizine, and all of them have glucuronide conjugates [30]. Intestinal bacterial
flora takes role in enterohepatic circulation of berberine and its conjugated
metabolites [28]. On the other hand, very small amount of unchanged berberine is
eliminated in urines [31].
As other alkaloids are present in H. canadensis extracts (i.e. hydrastine and
canadine), berberine may inhibit cytochrome P450 2E1 (CYP2E1) [32] and 1A2
(CYP1A2) [33]. This inhibition is not related to a significant increase in pharma-
cological interactions since the largest part of the available drugs is not metabolized
by these enzymatic systems.
kinase C (PKC) and nuclear factor-κB (NF-κB), interfering with the insulin sig-
nalling pathway and causing insulin resistance [39–41]. In addition, oxidative stress
also contributes to the development of chronic complications of diabetes, such as
diabetic nephropathy, retinopathy and neuropathy [37].
Molecular mechanisms of berberine in reducing oxidative stress seem to be
related with multiple cellular pathways (Fig. 2.1).
The NOX family of ROS-generating NADPH oxidases, a family of
membrane-associated enzymatic complexes, is one of the major sources of ROS
production in cells [42]; its activation is often associated to high levels of fatty
acids, cholesterol, glucose or advanced glycation end products (AGEs) [43–45].
Among various NOX isoforms, berberine was reported to suppress the overex-
pression of NOX 2,4 and to decrease ROS production in macrophages and
endothelial cells upon stimulation with inflammatory stimuli [46, 47]. In endothelial
cells, berberine attenuated LDL oxidation induced by ROS and reduces the collapse
of mitochondrial membrane potential, the chromosome condensation, the cyto-
chrome C release and the caspase-3 activation [48]. Circulating endothelial
BERBERINE
Nrf2 NF -kB AP -1
1 2 Nuclear
3 activation pathway
translocation inhibition
ROS Production
Inflammation
Oxidative stress
Fig. 2.1 Schematic illustration of the molecular mechanisms and pathways of Berberine in
reducing oxidative stress and inflammation. 1 Berberine could inhibit oxidative stress by
upregulation of SOD, and downregulation of NADPH oxidase expression. 2 Berberine
administration induces the activation of the Nrf2 transcription. The effect of berberine on Nrf2
relies on the activation of AMPK, and P38 pathways. 3 Berberine could suppress inflammation by
blocking the MAPK pathways in an AMPK-dependent manner, inhibiting the classic NF-κB
transcription; inhibiting the Rho GTPase pathway, which plays a role in NF-κB regulation, and
attenuating the transcription activity of AP-1, which was possible to be mediated by PPARγ
activation
2 Berberine and Its Role in Chronic Disease 31
[87, 88]. In insulin-resistant 3T3-L1 adipocytes [89] and liver/adipose tissues from
obese mice feed with HFD [74], berberine administration greatly reduced phos-
phorylation of ser181 and activation of IKK-β. In addition, the inhibitory effect of
berberine on IKK-β required a cysteine residue at position 179 of IKK-β [89].
Recent studies proved that berberine could reduce renal inflammation in diabetic
rats through inhibiting the Rho GTPase signalling pathway [69]. Rho GTPase is a
member of the superfamily of small GTP binding proteins with multiple biological
functions [90]; it was proven to positively regulate the NF-κB signalling pathway in
diabetic rats [91]. Therefore, in addition to regulation of the classic NF-κB sig-
nalling pathway, berberine could inhibit NF-κB by suppressing Rho GTPase [69,
92]. Furthermore, the inhibitory effect of berberine on Rho GTPase relied on its
antioxidant activity [69].
In addition to NF-κB, transcription factor activator protein 1 (AP-1) also played
a role in the anti-inflammatory activity of berberine [93, 94]. Administration of
berberine to macrophages or epithelial cells greatly attenuated the DNA binding
activity of AP-1 and reduced the production of cytokines like MCP-1 and COX2
[93]. There were reports that the transcription stimulating activity of AP-1 and
NF-κB could be inhibited by activation of peroxisome proliferator-activated
receptor γ (PPARγ) [95–99].
In general, there are two distinct pathways to activate glucose uptake in peripheral
tissues; one stimulated by insulin through the IRS-1/PI 3-kinase and the other by
exercise or hypoxia via activation of AMP activated protein kinase (AMPK). In
muscle, which is the major tissue responsible for whole body glucose disposal after
liver, both pathways stimulate the translocation of glucose transporter-4 (GLUT4)
to the cell membrane which accounts for the enhanced glucose uptake [100].
Current data suggest that the berberine effects are complex and may activate
portions of both the insulin and the exercise-induced glucose uptake pathways
[101]. In addition, berberine inhibits intestinal absorption of glucose, which also
contributes to berberine glucose-lowering effect [102].
There is increasing evidence that the most widely expressed GLUT1, initially
thought to be responsible only for basal glucose uptake, can be acutely activated by
cell stressors such as azide [103, 104], osmotic stress [105, 106], methylene blue [107]
and glucose deprivation [108, 109]. In particular, the acute activation of GLUT1 by
hypoxia or azide has been attributed to activation of AMPK [110, 111]. In addition, it
has been recently shown that peptide C activates GLUT1 transport activity in ery-
throcytes, establishing a potential link between GLUT1 activity and diabetes [112].
In cultured human liver cells and rat skeletal muscle, berberine increases insulin
receptor mRNA expression through Protein kinase C-dependent activation of its
promoter [113].
Since berberine was observed to act as an insulin-sensitizing agent in cultured
cells [114], its activity has been compared with that of metformin in different animal
34 A.F.G. Cicero and A. Baggioni
models. In rat models of type 2 diabetes (T2DM), berberine shows to have equal or
better fasting plasma glucose (FPG), insulin-resistance and low-density lipoprotein
cholesterol (LDL-C) lowering activity than metformin by a mechanism involving
retinol binding protein-4 (RBP-4) and (GLUT-4) [115, 116].
Berberine exhibited a high hypoglycemic potential; it has been shown that
berberine activates AMPK with subsequent induction of glycolysis [117]. AMPK,
as an intracellular energy receptor, has attracted more attention and become a new
target for the treatment of diabetes and its cardiovascular complications due to its
regulatory effect on endothelial cell function and energy homeostasis. In H9c2
myoblast cell line treated with insulin to induce insulin resistance, berberine
attenuated the reduction in glucose consumption and glucose uptake at least in part
via stimulation of AMPK activity [118]. berberine enhanced acute insulin-mediated
GLUT4 translocation and glucose transport in insulin-resistant myotubes through
activation of AMPK and PI3K pathway [119] (Fig. 2.2).
Berberine
Pancreas
β-cell
Insulin
receptor (IR)
Peripheral
IR
cells dephosphorylation
AMPK
Pathway
activation
Fig. 2.2 Main glucose-lowering effects of berberine in the human cells. Berberine administration
could decrease glycemia through the GLP-1 receptor activation in pancreas beta cells, the increase
of Insulin Receptor expression and the AMPK-modulated Glut-4 translocation in peripheral cells
2 Berberine and Its Role in Chronic Disease 35
In a clinical study, the same group observed that berberine significantly lowered
FPG, hemoglobin A1c, triglycerides and insulin levels in patients with T2DM as
well as metformin and rosiglitazone (a combination commonly used for the T2DM
therapy); the percentages of peripheral blood lymphocytes expressing InsR were
significantly elevated after therapy [120].
In a recent meta-analysis of randomized clinical trials, berberine resulted to be
safe and effective in the treatment of patients with T2DM [121].
The cholesterol and triglycerides lowering effect of berberine has been clearly
demonstrated by a recent meta-analysis of randomized clinical trials [122]. The
lipid-lowering activity of berberine, in association with other nutraceuticals, has
been also clearly confirmed in a relatively large number of randomized clinical
trials [123, 124].
The supposed mechanism of action is the increased expression of the liver
receptor for LDL mediated by the inhibition of the Pro-protein-convertase-
subtilisin-kexin-9 (PCSK9) activity [125]. Besides its upregulation effect on the
LDL receptor, berberine could also reduce triglycerides by AMP kinase activation
and MAPK/ERK pathway blocking [126] (Fig. 2.3).
High levels of LDL and their oxidized counterpart, oxidized LDL (oxLDL), in
the blood vessels represent a major risk factor for endothelial dysfunction and
atherosclerosis [127]. Inactivity of LDL receptor (LDLR) or its low-level expres-
sion initiates accumulation of LDL in blood vessels [128]. On the other hand, the
receptor of oxLDL, lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1)
identified as the main endothelial receptor for oxLDL also present in macrophages
and smooth muscle cells (SMC), activates a proatherogenic cascade by inducing
endothelial dysfunction, SMC proliferation, apoptosis and the transformation of
macrophages into foam cells and platelet activation via NF-κB activation [129].
LOX-1 contains a lectin-like extracellular C-terminal domain which interacts with
oxLDL, proteolytically cleaved and released as a soluble circulating form (sLOX-1)
that reflects the increased expression of membrane-bound receptors and disease
activities [130].
In human macrophage-derived foam cells treated with oxLDL, berberine inhibits
the expression of LOX-1 [131] as well as the oxLDL uptake of macrophages and
reduces foam cell formation in a dose-dependent manner [132] by activating the
AMPK-SIRT1-PPARγ pathway [133]. Chi and colleagues demonstrated that ber-
berine combined with atorvastatin is more effective in diminishing LOX 1
expression than atorvastatin alone in monocyte-derived macrophages both in vitro
and in rats through modulation of endothelin-1 receptor [134].
Berberine improves also the survival of TNFα-treated endothelial progenitor
cells (EPCs) via the activation of PI3K/AKT/eNOS transcription factor [135]
possibly through AMPK activation. Wu and colleagues showed, both in vitro and
in vivo that berberine reduces the leukocyte-endothelium adhesion and vascular cell
36 A.F.G. Cicero and A. Baggioni
LDL- C decrease
LDL- C receptors
Liver cell
membrane
TG decrease
LDL- C receptors
upregulation
MAPK/ERK inhibition
PCSK9 Inhibition
AMPK activation
BERBERINE
Fig. 2.3 Main lipid-lowering effects of berberine in the human liver cells. Berberine mainly
decreases circulating LDLs by inducing LDLR expression in hepatic cells mediated by inhibition
of PCSK9
Highly purified and concentrated berberine is safe, in fact, its Lethal Dose 50
(LD50) in mice is 25 mg/kg in mice [131].
Standard doses of berberine are usually well tolerated and adverse events are rare
and mild. The most studied side effects are those on the gastrointestinal system. In
fact, berberine and its derivatives can produce gastric lesions in animal models
[142]. As shown by the determination of small intestinal transit time measurements
by sorbitol and breath hydrogen test, berberine delays small intestinal transit time,
and this may account for a part of its gastrointestinal side effects (but also of its
antidiarrhoeal one) [143].
The main safety issue of berberine involves the risk of pharmacological inter-
actions. In fact, berberine displaces bilirubin from albumin about tenfold more than
phenylbutazone, thus any herb containing large amounts of berberine should be
avoided in jaundiced infants and pregnant women [144]. Berberine also displaces
warfarin, thiopental and tolbutamide from their protein binding sites, increasing
their plasma levels [145].
Then, berberine can markedly increase blood levels of cyclosporine A because
of CYP3A4 and P-glycoprotein inhibition in liver and gut wall, respectively, and
because of the increase in gastric emptying time, thus causing increased cyclos-
porine A bioavailability and reduced metabolism [146]. In renal transplant recipi-
ents who take cyclosporine 3 mg/kg twice daily, the coadministration of berberine
(0.2 g/day for three times a day for 3 months) increased the mean cyclosporine A
AUC of 34.5 % and its mean half-life of 2.7 h [147].
Even if the main mechanism of berberine pharmacological interaction involves
CYP3A4 and intestinal P-glycoprotein, it also inhibits CYP1A1, potentially inter-
acting with drugs metabolized by this cytochrome isophorm as well. The impact of
this observation in clinical practise has yet to be evaluated since the CYP1A1
metabolized drugs are relatively rare [148].
Overall, the assumption of berberine in dosages of 500–1000 mg/day has to be
considered safe for the most part of subjects and the risk of clinically relevant
pharmacological interaction is limited to cyclosporine and warfarin.
38 A.F.G. Cicero and A. Baggioni
2.2 Conclusion
References
16. Timothy CBN, Gregory S, Kelly ND (1997) Berberine: therapeutic potential of an alkaloid
found in several medicinal plants. Altern Med Rev 13:94–103
17. Kunwar RM, Nepal BK, Kshhetri HB et al (2006) Ethnomedicine in Himalaya: a case study
from Dolpa, Humla, Jumla and Mustang districts of Nepal. J Ethnobiol Ethnomed 13:27
18. Fatehi-Hassanabad Z, Jafarzadeh M, Tarhini A, Fatehi M (2005) The antihypertensive and
vasodilator effects of aqueous extract from Berberis vulgaris fruit on hypertensive rats.
Phytother Res 13(3):222–225
19. Ivanovska N, Philipov S (1996) Study on the anti-inflammatory action of Berberis vulgaris
root extract, alkaloid fractions and pure alkaloids. Int J Immunopharmacol 13(10):553–561
20. Imanshahidi M, Hosseinzadeh H (2008) Pharmacological and therapeutic effects of Berberis
vulgaris and its active constituent, Berberine. Phytother Res 13:999–1012
21. Mills S, Bone K (2000) Principals and practice of phytotherapy. Churchill Livingstone,
Edinburgh, pp 338–341
22. Bone K (2003) A clinical guide to blending liquid herbs: herbal formulations for the
individual patient. Churchill Livingstone, St Louis, pp 422–429
23. Khosrokhavar R, Ahmadiani AS (2010) Antihistaminic and anticholinergic activity of
methanolic extract of barberry fruit (Berberis vulgaris) in the Guinea-Pig Ileum. J Med Plants
13:99–105
24. Akhter MH, Sabir M, Bhide NK (1979) Possible mechanism of antidiarrheal effect of
berberine. Indian J Med Res 13:233–241
25. Rabbani GHBT, Knight J, Sanyal SC, Alam K (1987) Randomized controlled trial of
berberine sulfate therapy for diarrhea due to enterotoxigenic Escherichia coli and Vibrio
cholera. J Infect Dis 13:979–984
26. Amin AH, Subbaiah TV, Abbasi KM (1969) Berberine sulfate: antimidrobial activity,
bioassay, and mode of action. Can J Microbiol 13:1067–1076
27. Sabir M (1971) Study of some pharmacological actions of berberine. Indian J Physiol
Pharmacol 13:111–132
28. Pan GY, Wang GJ, Liu XD, Fawcett JP, Xie YY (2002) The involvement of P-glycoprotein
in berberine absorption. Pharmacol Toxicol 91:193–197
29. Tsai PL, Tsai TH (2003) Hepatobiliary excretion of berberine. Drug Metab Dispos 32:405–
412
30. Zuo F, Nakamura N, Akao T, Hattori M (2006) Pharmacokinetics of berberine and its main
metabolites in conventional and pseudo germ-free rats determined by liquid
chromatography/ion trap mass spectrometry. Drug Metab Dispos 34:2064–2072
31. Chen CM, Chang HC (1995) Determination of berberine in plasma, urine and bile by high
performance liquid chromatograpy. J Chromatogr 665:117–123
32. Raner GM, Cornelious S, Moulick K et al (2007) Effects of herbal products on human
cytochrome P450(2E1) activity. Food Chem Tox 45:2359–2365
33. Zhao X, Zhang JJ, Wang X et al (2008) Effect of berberine on hepatocyte proliferation
inducible nitric oxide synthase expression, cytochrome 450 2E1 and 1A2 activities in
diethylnitrosamine- and Phenobarbital-treated rats. Biomed Pharmacother 62:567–572
34. Furukawa S, Fujita T, Shimabukuro M et al (2004) Increased oxidative stress in obesity and
its impact on metabolic syndrome. J Clin Investig 114(12):1752–1761
35. Bonnefont-Rousselot D (2002) Glucose and reactive oxygen species. Curr Opin Clin Nutr
Metab Care 5(5):561–568
36. Alberici LC, Vercesi AE, Oliveira HC (2011) Mitochondrial energy metabolism and redox
responses to hypertriglyceridemia. J Bioenerg Biomembr 43(1):19–23
37. Rösen P, Nawroth PP, King G et al (2001) The role of oxidative stress in the onset and
progression of diabetes and its complications: a summary of a congress series sponsored by
UNESCO-MCBN, the American diabetes association and the German diabetes society. Diab
Metab Res Rev 17(3):189–212
38. Evans JL, Goldfine ID, Maddux BA, Grodsky GM (2003) Are oxidative stress—activated
signaling pathways mediators of insulin resistance and β-cell dysfunction? Diabetes 52(1):1–
8
40 A.F.G. Cicero and A. Baggioni
76. Xing L-J, Zhang L, Liu T (2011) Berberine reducing insulin resistance by up-regulating
IRS-2 mRNA expression in nonalcoholic fatty liver disease (NAFLD) rat liver. Eur J
Pharmacol 668(3):467–471
77. Cui G, Qin X, Zhang Y et al (2009) Berberine differentially modulates the activities of ERK,
p 38 MAPK, and JNK to suppress Th17 and Th1 T cell differentiation in type 1 diabetic
mice. J Biol Chem 284(41):28420–28429
78. Chueh WH, Lin JY (2012) Protective effect of isoquinoline alkaloid berberine on
spontaneous inflammation in the spleen, liver and kidney of non-obese diabetic mice
through downregulating gene expression ratios of pro-/anti-inflammatory and Th1/Th2
cytokines. Food Chem 131(4):1263–1271
79. Zhang Y, Li X, Zou D et al (2008) Treatment of type 2 diabetes and dyslipidemia with the
natural plant alkaloid berberine. J Clin Endocrinol Metab 93(7):2559–2565
80. Meng S, Wang LS, Huang ZQ et al (2012) Berberine ameliorates inflammation in patients
with acute coronary syndrome following percutaneous coronary intervention. Clin Exp
Pharmacol Physiol 39(5):406–411
81. Dubois RN, Abramson SB, Crofford L (1998) Cyclooxygenase in biology and disease.
FASEB J 12(12):1063–1073
82. Mima A (2013) Inflammation and oxidative stress in diabetic nephropathy: new insights on
its inhibition as new therapeutic targets. J Diab Res 2013:248563
83. Lee D, Bae J, Kim YK et al (2013) Inhibitory effects of berberine on
lipopolysaccharide-induced inducible nitric oxide synthase and the high-mobility group
box 1 release in macrophages. Biochem Biophys Res Commun 431(3):506–511
84. Zhou Y, Liu SQ, Yu L et al (2015) Berberine prevents nitric oxide-induced rat chondrocyte
apoptosis and cartilage degeneration in a rat osteoarthritis model via AMPK and p38 MAPK
signaling. Apoptosis 20(9):1187–1199
85. Gratas-Delamarche A, Derbré F, Vincent S, Cillard J (2014) Physical inactivity, insulin
resistance, and the oxidative-inflammatory loop. Free Rad Res 48(1):93–108
86. Solinas G, Karin M (2010) JNK1 and IKKbeta: molecular links between obesity and
metabolic dysfunction. FASEB J 24(8):2596–2611
87. Karin M (1999) Positive and negative regulation of IκB kinase activity through IKKβ subunit
phosphorylation. Science 284(5412):309–313
88. Yi P, Lu FE, Xu LJ et al (2008) Berberine reverses free-fatty-acid-induced insulin resistance
in 3T3-L1 adipocytes through targeting IKKβ. World J Gastroenterol 14(6):876–883
89. Pandey MK, Sung B, Kunnumakkara AB et al (2008) Berberine modifies cysteine 179 of
IκBα kinase, suppresses nuclear factor-κB-regulated antiapoptotic gene products, and
potentiates apoptosis. Cancer Res 68(13):5370–5379
90. Shi J, Wei L (2013) Rho kinases in cardiovascular physiology and pathophysiology: the
effect of fasudil. J Cardiovasc Pharmacol 62(4):341–354
91. Xie X, Peng J, Chang X et al (2013) Activation of RhoA/ROCK regulates NF-κB signaling
pathway in experimental diabetic nephropathy. Mol Cell Endocrinol 369(1–2):86–97
92. Remppis A, Bea F, Greten HJ et al (2010) Rhizoma Coptidis inhibits LPS-induced
MCP-1/CCL2 production in murine macrophages via an AP-1 and NFκB-dependent
pathway. Mediat Inflamm 2010:194896
93. Kuo CL, Chi CW, Liu TY (2004) The anti-inflammatory potential of berberine in vitro and
in vivo. Cancer Lett 203(2):127–137
94. Schonthaler HB, Guinea-Viniegra J, Wagner EF (2011) Targeting inflammation by
modulating the Jun/AP-1 pathway. Ann Rheum Dis 70(1):i109–i112
95. Ricote M, Li AC, Willson TM et al (1998) The peroxisome proliferator-activated receptor-γ
is a negative regulator of macrophage activation. Nature 391(6662):79–82
96. Delerive P, Martin-Nizard F, Chinetti G et al (1999) Peroxisome proliferator-activated
receptor activators inhibit thrombin-induced endothelin-1 production in human vascular
endothelial cells by inhibiting the activator protein-1 signaling pathway. Circ Res 85(5):394–
402
2 Berberine and Its Role in Chronic Disease 43
97. Pasceri V, Wu HD, Willerson JT, Yeh ETH (2000) Modulation of vascular inflammation
in vitro and in vivo by peroxisome proliferator-activated receptor-γ activators. Circulation
101(3):235–238
98. Chen FL, Yang ZH, Liu Y et al (2008) Berberine inhibits the expression of TNFα, MCP-1,
and IL-6 in AcLDL-stimulated macrophages through PPARγ pathway. Endocrine 33(3):331–
337
99. Feng AW, Gao W, Zhou GR et al (2012) Berberine ameliorates COX-2 expression in rat
small intestinal mucosa partially through PPARγ pathway during acute endotoxemia. Int
Immunopharmacol 12(1):182–188
100. Krook A, Wallberg-Henriksson H, Zierath JR (2004) Sending the signal: molecular
mechanisms regulating glucose uptake. Med Sci Sports Exerc 36:1212–1217
101. Kim SH, Shin EJ, Kim ED (2007) Berberine activates GLUT1-mediated glucose uptake in
3T3-L1 adipocytes. Biol Pharm Bull 30:2120–2125
102. Pan GY, Huang ZJ, Wang GJ et al (2003) The antihyperglycaemic activity of berberine arises
from a decrease of glucose absorption. Planta Med 69:632–636
103. Shetty M, Loeb JN, Vikstrom K, Ismail-Beigi F (1993) Rapid activation of GLUT-1 glucose
transporter following inhibition of oxidative phosphorylation in clone 9 cells. J Biol Chem
268:17225–17232
104. Rubin D, Ismail-Beigi F (2003) Distribution of Glut1 in detergent-resistant membranes
(DRMs) and non-DRM domains: effect of treatment with azide. Am J Physiol Cell Physiol
285:C377–C383
105. Barnes K, Ingram JC, Porras OH et al (2002) Activation of GLUT1 by metabolic and
osmotic stress: potential involvement of AMP-activated protein kinase (AMPK). J Cell Sci
115:2433–2442
106. Barros LF, Barnes K, Ingram JC et al (2001) Hyperosmotic shock induces both activation
and translocation of glucose transporters in mammalian cells. Pflugers Arch 442:614–621
107. Louters LL, Dyste SG, Frieswyk D et al (2006) Methylene blue stimulates 2-deoxyglucose
uptake in L929 fibroblast cells. Life Sci 78:586–591
108. Kumar A, Xiao YP, Laipis PJ (2004) Glucose deprivation enhances targeting of GLUT1 to
lipid rafts in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 286:E568–E576
109. Roelofs B, Tidball A, Lindborg AE et al (2006) Acute activation of glucose uptake by
glucose deprivation in L929 fibroblast cells. Biochimie 88:1941–1946
110. Jing M, Ismail-Beigi F (2007) Critical role of 5′-AMP-activated protein kinase in the
stimulation of glucose transport in response to inhibition of oxidative phosphorylation. Am J
Physiol Cell Physiol 292:C477–C487
111. Jing M, Cheruvu VK, Ismail-Beigi F (2008) Stimulation of glucose transport in response to
activation of distinct AMPK signaling pathways. Am J Physiol Cell Physiol 295:C1071–
C1082
112. Meyer JA, Froelich JM, Reid GE, Karunarathne WK, Spence DM (2008) Metal-activated
C-peptide facilitates glucose clearance and the release of a nitric oxide stimulus via the
GLUT1 transporter. Diabetologia 51:175–182
113. Kong WJ, Zhang H, Song DQ et al (2009) Berberine reduces insulin resistance through
protein kinase C-dependent up-regulation of insulin receptor expression. Metabolism
58:109–119
114. Ko BS, Choi SB, Park SK et al (2005) Insulin sensitizing and insulin otropic action of
berberine from Cortidis rhizoma. Biol Pharm Bull 28:1431–1437
115. Zhang W, Xu YC, Guo FJ, Meng Y, Li ML (2008) Antidiabetic effects of cinnamaldehyde
and berberine and their impacts on retinol binding protein 4 expression in rats with type 2
diabetes mellitus. Chin Med J121:2124–2128
116. Ni WJ, Ding HH, Tang LQ (2015) Berberine as a promising anti-diabetic nephropathy drug:
An analysis of its effects and mechanisms. Eur J Pharmacol 760:103–112
117. Yin J, Gao Z, Liu D, Liu Z, Ye J (2008) Berberine improves glucose metabolism through
induction of glycolysis. Am J Physiol Endocrinol Metab 294:E148–E156
44 A.F.G. Cicero and A. Baggioni
139. Peng WH, Lo KL, Lee YH, Hung TH, Lin YC (2007) Berberine produces antidepressant-like
effects in the forced swim test and in the tail suspension test in mice. Life Sci 81:933–938
140. Kong LD, Cheng CH, Tan RX (2001) Monoamine oxidase inhibitors from rhizoma of Coptis
chinensis. Planta Med 67:74–76
141. Kumar A, Ekavali Chopra K et al (2015) Current knowledge and pharmacological profile of
berberine: an update. Eur J Pharmacol 761:288–297
142. Kupeli E, Kosar M, Yesilada E, Hüsnü K, Başer C (2002) A comparative study on the
anti-inflammatory, antinociceptive and antipyretic effects of isoquinoline alkaloids from the
roots of Turkish berberis species. Life Sci 72:645–657
143. Chen C, Yu Z, Li Y, Fichna J, Storr M (2014) Effects of berberine in the gastrointestinal tract
—a review of actions and therapeutic implications. Am J Chin Med 42(5):1053–1070
144. Chan E (1993) Displacement of bilirubin from albumin by berberine. Biol Neonat 63:201–
208
145. Tan YZ, Wu AC, Tan BY et al (2002) Study on the interactions of berberine displace other
drug from their plasma proteins binding sites. Chin Pharmacol Bull 18:576–578
146. Xin HW, Wu XC, Li Q et al (2006) The effects of berberine on the pharmacokinetics of
cyclosporine A in healthy volunteers. Methods Find Exp Clin Pharmacol 28:25–29
147. Wu X, Lu Q, Xin H, Zhong M (2005) Effects of berberine on the blood concentration of
cyclosporine A in renal transplanted recipients: clinical and pharmacokinetic study. Eur J
Clin Pharmacol 61:567–572
148. Cicero AF, Tartagni E, Ertek S (2014) Safety and tolerability of injectable lipid-lowering
drugs: a review of available clinical data. Expert Opin Drug Saf 13(8):1023–1030
Chapter 3
Emodin and Its Role in Chronic Diseases
Abstract Diseases, such as heart disease, stroke, cancer, respiratory diseases, and
diabetes, are by far the leading cause of mortality in the world, representing 60 % of
all deaths. Although substantial medical advances have been made and many
therapeutic approaches proposed yet traditional medicine and medicinal plants find
an important place in therapy. They have been providing invaluable solutions to the
various health problems. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is a
natural anthraquinone derivative found in various Chinese medicinal herbs.
Traditionally, it has been used as an active constituent of many herbal laxatives.
However, in the last few years, significant progress has been made in studying the
biological effects of emodin at cellular and molecular levels and it is emerging as an
important therapeutic agent. This review provides an overview of the modulatory
effects of emodin in various diseases and cell signaling pathways, which may have
important implications in its future clinical use.
Keywords Emodin Bioavailability Cancer Anthraquinone Radiosensitizer
Chemosesitizer
3.1 Introduction
B.A. Monisha
Department of Biomedical Science, Bharathidasan University,
Tiruchirappalli, Tamil Nadu 620024, India
N. Kumar A.B. Tiku (&)
Radiation and Cancer Therapeutics Lab, School of Life Sciences,
Jawaharlal Nehru University, New Delhi 110067, India
e-mail: [email protected]
in higher plants and in isolated instances in insects [24, 131]. Among higher plants,
plants belonging to families Rubiaceae, Rhamnaceae, Fabaceae, Polygonaceae,
Bignoniaceae, Verbenaceae, Scrophulariaceae, and Liliaceae are rich sources of
anthraquinones [134]. Emodin, rhein, chrysophanol, aloe-emodin, and physcion are
the most common naturally occurring anthraquinonesin higher plants [24, 33].
The anthraquinone emodin is identified in 17 plant families distributed world-
wide but is primarily reported in three plant species Fabaceae (Cassia spp.),
Polygonaceae (Rheum, Rumex, and Polygonum spp.), and Rhamnaceae (Rhamnus
and Ventilago spp.) [43]. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is
present in bark, root, vegetative organ (stem, foliage), reproductive organ (flower,
fruit, seeds, pods), and is produced as secondary metabolite by molds and lichens
[120].
Although emodin was first described more than 75 years ago (reported as
‘frangula-emodin,’ [56], many of its diverse biological properties have been dis-
covered in the last decade (see reviews by Srinivas et al. [120] and Shrimali et al.
[117]). Emodin has also been reported to play a significant ecological role in the life
of many plant species by mediating their interactions with their biotic and abiotic
environment [43].
Emodin is a bioactive anthraquinone and has been an active constituent of many
laxatives and Chinese herbal medicines [71, 73]. It has antitumour, antibacterial,
diuretic, and vasorelaxant effects [39, 57, 175]. It induces growth inhibition in
cancer cells but not in normal cells [98, 122, 162] and modulates cellular redox
status in a dose- and time-dependent manner [58, 120, 163]. The photo-protective
function of emodin against ultraviolet (UV) region of the solar radiation (290–
400 nm) has also been reported [8]. Protective role of emodin against
radiation-induced oxidative and DNA damage in murine splenocytes and in the
concanavalin A (ConA)-induced hyperproliferation was also reported [109, 110]. It
possesses immunosuppressive activities also [85, 142, 145]. Various pharmaco-
logical properties of emodin in both animal and human model systems have been
tabulated (Tables 3.1, 3.2).
Emodin is highly effective in case of pancreatitis, asthma, myocarditis, arthritis,
atherosclerosis, glomerulonephritis, Alzheimer’s, hepatitis, and chronic obstructive
lung disease [117, 156, 157]. It modulates various signaling pathways and produces
many therapeutic effects (Table 3.3).
Besides the beneficial effects, emodin has been reported to cause some toxic side
effects, such as genotoxicity, developmental toxicity, nausea, diarrhea, and renal
failure. Recently, Sevcovicova et al. [108] showed emodin exhibited dual activities;
on one side it was genotoxic inducing primary DNA lesions as well as gene
mutations and on the other it exhibited DNA-protective activity via free radicals
scavenging and reducing activities. Therefore, safety and effectiveness of emodin in
naturopathic treatment is yet to be approved by the U.S. Food and Drug
Administration (FDA). The present review is the compilation of the literature on
effects of emodin in various disease conditions and the underlying molecular
mechanisms.
3 Emodin and Its Role in Chronic Diseases 49
Table 3.3 Therapeutic effects of emodin in chronic diseases and mechanisms of action
Diseases Mechanism References
Allergy Inhibits TNF-α secretion through Lu et al. [88], Kim et al. [53],
the inhibition of PKC or Nemmar et al. [99]
PKC-IKK2 pathways
Cancer Targets PI3K/Akt pathway Su et al. [121], Olsen et al. [103],
Downregulates TGF-β signaling Hsu et al. [37], Yan et al. [158],
pathway and Inhibits β-catenin/Akt Way et al. (2014), Thacker and
Downregulates cytoprotective ERK Karunagaran [129, 130], Li et al.
and Akt cascade [77], Deng et al. [19], Hu et al.
Elevates the levels of Bax, reduces [38], Tang et al. [128], Sun et al.
Bcl-2 and activates caspase-2, -3, [124]
and -9, Suppresses the activation of
P210BCR−ABL downstream
signaling pathways including
CrkL, Akt/mTOR and MEK/ERK
Inhibits TOR signaling pathway
and blocks autophagy
Inhibits ILK expression through
AMPKα-mediated reduction of Sp1
and c-Jun proteins and suppresses
the activation of MAPK signaling
pathways
Inhibits Wnt signaling pathway like
(i) regulating the regulators-p300
and HBP1 (ii) increasing reactive
oxygen species
Cardiovascular Enhances mitochondrial Du et al. [23], Wu et al. [149],
diseases antioxidant components and Song et al. [119], Chen et al.
inducesTNF-α upregulation and [10, 12, 14], Jiang et al. [46]
cardiomyocyte apoptosis
Suppresses pro-inflammatory
cytokines TNF-α and IL-1β due to
inhibition of NF-κB activation
Inhibits IL-23/IL-17 inflammatory
axis, Th17cell proliferation and
viral replication mRNA/protein
Diabetes Regulates PPAR-γ and Xue et al. [155], Wang et al. [142,
AKT/GSK-3β signaling pathway 145], Wu et al. [148, 150],
Arvindekar et al. [3]
Kidney Activates autophagy by modulating Gao et al. [27], Liu et al. [81]
diseases AMPK/mTOR signaling pathways
Liver diseases Increases the mRNA levels of Zhan et al. [167], Dong et al. [20],
PPAR-γ Meng et al. [94], Dong et al. [22],
Inhibits p38 MAPK-NF-κB Lin et al. [81], Dang et al. [18],
pathway leading to suppression of Dong et al. [22], Lee et al. [61],
hepatic IFN-γ, TNF-α, IL-1β, Tzeng et al. [133], Liu et al. [84],
IL-12, IL-6, iNOS, ITGAM, CCL2, Xue et al. [156]
and MIP-2, MIP-2 receptor, and
CXCR2
(continued)
54 B.A. Monisha et al.
the keto forms can be considered negligible for emodin as a free molecule. It is clear
that emodin appears to be a planar molecule and the most stable radical in the gas
phase is the 3-OH species. Despite planarity of radical structures, there is no sig-
nificant electronic delocalisation between adjacent rings [92].
Emodin–DNA interaction mainly involves intercalation of emodin between
bases and with PO2 backbone. Interaction occurs mainly through Ade and Thy
bases and PO2 backbone of double helix. Binding constant for emodin–DNA is
5.59 × 10−3 M−1 [107]. Emodin has low DNA-binding affinity and has low
cytotoxicity against various cancer cells. Addition of pyrazole ring and certain
chemical groups like polymethyleneamine, sugar with anthraquinone chromophore
result in increased binding affinity and cytotoxicity against various cancers. Chain
of varying length, polarity, charge, rigidity, and steric bulk may impart different
DNA binding affinity. Emodin with mono-cationic amino side chain has stronger
cytotoxic potential against cancer cells than di-cationic amino side chain, as indi-
cated by cytotoxicity potency index (IC50) value [154].
Absorption, excretion, tissue distribution, and metabolism of emodin were
studied after a single oral administration of C14-labeled emodin (50 mg/kg) in rat
model [4]. Emodin was quickly absorbed from the gastrointestinal tract.
Radioactivity in the peripheral blood reached a peak 2 h after administration, and
within 24 h subsequently decreased to 30 % of the peak value. In two cannulated
rats, biliary excretion reached a maximum at approximately 6 h and amounted to
49 % dose within 15 h; 70 % of biliary activity was in the form of conjugated
emodin. Urinary excretion amounted to 18 and 22 % dose, in 24 and 72 h,
respectively and most metabolites in pooled urine found were free emodin and
emodic acid. Emodin glycosides are carried unabsorbed to the large intestine
(because of its chemical structure) where metabolism to the active aglycone takes
place by intestinal bacterial flora. The aglycone damages epithelial cells, which
leads directly and indirectly to changes in absorption, secretion, motility and exerts
its laxative effect [97, 135].
In Male Sprague-Dawley rats orally administered PC Polygonum cuspidatum a
widely used Chinese medicine which contains resveratrol and emodin, it was found
that the sulfates/glucuronides of resveratrol and emodin were the major forms in
circulation and in most assayed organs after oral intake [78, 79]. With regard to
tissue distribution emodin was detected as sulfates/glucuronides in lung and kidney,
as free form in liver, but was not detectable in brain and heart [113, 114]. In rats
poor oral bioavailability of emodin is thought to be the result of intestinal and
hepatic glucuronidation [83]. The activity and positional preference of glu-
curonidation of anthraquinones varies with organs, species, substrate concentra-
tions, UGT isoforms, and the substitution at b-positions [148, 150]. Generally, the
conjugated metabolites are recognized as the inactive product of drugs, however,
now there are increasing evidences showing that the conjugated metabolites of
polyphenols demonstrate various bioactivities [25, 111, 112, 116, 159, 166].
Emodin which mainly exists as conjugated metabolite in the circulation and in most
organs, needs to be extensively investigated in future.
56 B.A. Monisha et al.
3.3.1 Allergy
Emodin plays an important role in allergic diseases like asthma, rhinitis, and atopic
dermatitis [47, 88]. Asthma is a respiratory disease associated with symptoms like
airway hyper responsiveness, mucus hypersecretion, and bronchial inflammation
[139]. Emodin can be a therapeutic agent in treating allergic airway inflammation. It
inhibited ovalbumin-induced increase in eosinophil counts and hypersecretion of
mucus from goblet cells in air way passage [17]. Emodin also exhibited antiallergic
activities via increasing the stability of the cell membrane and inhibiting extracel-
lular Ca2+ influx [142, 145].
Mast cells play a major role in allergic diseases. Emodin lowered mast
cell-dependent passive anaphylactic reaction in Ig-E sensitized mice and inhibited
degranulation, generation of eicosanoid, and secretion of cytokines in
dose-dependent manner in mast cell [53, 88, 117]. Mast cell degranulation inhi-
bition occurred via attenuation of protein kinase C and IƙB kinase 2 signaling
pathway [53].
3.3.2 Arthritis
3.3.3 Cancer
Recent studies on emodin are mostly focused on its antitumor properties. These
efforts led to unraveling both the effect of emodin against cancer development and
the underlying molecular mechanisms involved. A number of studies have
demonstrated that emodin inhibited the growth and proliferation of various cancer
cells derived from different tumors, such as cervical, breast, lung, colorectal, and
prostate cancers (Tables 3.1, 3.2). After evaluation with other anthraquinone
derivatives, including emodin 1-O-β-D-glucoside, physcion 1-O-β-D-glucoside,
and physcion, C1 and C3 position of emodin was supposed to be important for its
antitumor function [59]. Meanwhile, emodin displayed over 25-fold differential
cytotoxicity against ras-transformed bronchial epithelial cells to the normal human
bronchial epithelial cells [6]. Emodin also evoked a less or no cytotoxic effect in
several normal cells, together with human fibroblast-like lung WI-38 cells,
HBL-100 cells derived from normal human breast tissue and three primary cultured
rat normal cells [115, 170]. Recently, Sharma and Tiku [109] reported noncytotoxic
effects on murine splenocytes up to 100 µM of emodin. These observations sug-
gested that normal cells might be more resistant to emodin-induced cytotoxicity
than cancer cells. Cells contain various pathways designed to protect them from the
genomic instability or toxicity that can result when their DNA is damaged.
A pivotal role in this response is played by checkpoint proteins that control the
normal passage of cells through the cell cycle. The effect of emodin on cell cycle
has been demonstrated on various cancer cells. In Her2/neu-over expressing
MDA-MB-453 breast cancer cells emodin azide methyl derivative (AMAD) trig-
gered mitochondrial-dependent cell apoptosis involving caspase-8-mediated Bid
cleavage. This derivative-induced G0/G1 arrest by blocking Her2/neu binding to
Hsp90. This was associated with decreasing protein expression of c-Myc, Cyclin
D1, CDK4, and p-Rb [123, 158, 169]. Emodin-induced G2/M phase arrest in
v-ras-transformed cells [6] and the human hepatoma cell line HepG2/C3A cells
[115]. Elevation of p53 and p21 expression might be involved in this G2/M arrest
[115]. In addition to G2/M phase arrest, emodin was reported to block the G1 to S
phase of the cell cycle in human colon carcinoma HCT-15 cells [50] and breast
cancer MDA-MB-453 cells [170]. It downregulated Wnt signaling pathway in
human colorectal cancer cells SW480 and SW620 [129]. Emodin attenuated
radioresistance in the HepG2 cells via upregulation of the apoptotic signals and
downregulation of the proliferative signals [42]. Emodin also inhibited the lung
metastasis of human breast cancer in a mouse xenograft model, and inhibited the
invasion of MDA-MB-231 cells associated with the downregulation of MMP-2,
MMP-9, uPAR, and uPA expression as well as decreased activity of p38 and ERK
[124]. Combination of emodin with curcumin synergistically inhibited survival,
proliferation, and invasion of breast cancer cells and cervical cancer [130].
Apoptosis could be a potential general mechanism of the anti-proliferative and
antineoplastic effects of emodin. A number of studies have demonstrated that
emodin is capable of inducing apoptotic cell death in various cancer cells [120].
58 B.A. Monisha et al.
(causal agent of viral myocarditis) by inhibiting CVB3 VP1 protein translation. The
fundamental signaling pathways involved in inhibition of CVB3 VP1 protein
translation include Akt-mTORC1-4EBP1, mTORC1-p70S6K, ERK1/2-p90RSK,
and Ca2+-calmodulin (Ca2+-CaM) cascades. Therefore, these pathways might be the
targets for emodin [168].
Atherosclerosis a chronic inflammatory disease characterized by the presence of
atherosclerotic plaque in the arterial intima can lead to hardening and narrowing of
the major arteries. Emodin inhibited NF-κB activation and TNF-α-induced migra-
tion, proliferation and MMP-2, MMP-9 expression in rat aortic smooth muscle cells
(RASMCs) [95]. It also had beneficial effects on stability of atherosclerotic plaque
in Apo-E deficient mice (apo-E has anti-atherosclerosis property) and was found to
inhibit the expression of M-CSF and MMP-9, whereas it enhanced the PPAR-γ
expression [174]. Cardiovascular protective actions of emodin have been attributed
to activation of cardiac natriuretic hormone secretion.
In isolated rabbit atria, emodin heighted atrial natriuretic peptide (ANP) secre-
tion via inhibition of L-type calcium channels and activation of potassium ATP
channels [176]. C-reactive protein (CRP) an inflammatory molecule plays a direct
role in atherogenesis. Emodin was also found to inhibit Hcy-induced CRP gener-
ation in vascular smooth muscles cells via ROS-ERK1/2/p38 signaling pathway and
upregulated PPARγ expression [104]. The above-mentioned results provide a
rationale for the use of emodin in the treatment of cardiovascular homeostasis and
other heart diseases.
3.3.5 Diabetes
level was also significantly reduced in the liver and adipose tissue after emodin
treatment [69].
Emodin may also be a potential medication in treating the glucose dependent
structural and functional abnormalities in peritoneal membrane. Emodin regulated
the undesirable effects of concentrated glucose on human peritoneal mesothelial
cells by suppression of protein kinase C (PKC) activation and cyclic AMP response
element binding protein (CREB) phosphorylation [7]. Emodin was found to protect
against diabetic cardiomyopathy by regulating AKT/GSK-3β signaling pathway
[148, 150]. In the presence of high-glucose concentration human umbilical vein
endothelial cells (HUVECs) cultured with 3 µm of emodin showed protection from
endothelial cytotoxicity by suppressing MAPK pathway and inhibiting chemokine
ligand 5 (CCL5) expression [30].
Autoimmune diabetes (AID) is a metabolic disease that progresses through an
intricate relationship of environmental, genetic, and immune factor. Emodin was
able to suppress the chemotactic activity of leukocytes at the insulitis stage
(inflammation of the islets of Langerhans) of AID development [15].
Emodin was also found to have beneficial effects in renal dysfunction. In diabetic
nephropathy it inhibited activation of p38 MAPK pathway and downregulated the
expression of fibronectin. This effect was independent of the blood glucose level
[137]. Emodin was able to protect mice from drug-induced kidney injury. In
cyclosporine-induced kidney nephropathy emodin prevented the overexpression of
Protein Kinase Casein Kinase II (PKCK2) which has a role in apoptosis [118].
In LPS treated NRK-52E cells (Rat kidney epithelial cells) it inhibited
TLR2-mediated NF-κB signaling pathway [70]. Emodin was found to ameliorate
cisplatin-induced apoptosis of rat renal tubular cells in vitro through modulating the
AMPK/mTOR signaling pathways and activating autophagy. Emodin may have
therapeutic potential for the prevention of drug-induced nephrotoxicity [81].
mice, partially through inhibition of both the infiltration of CD4(+) and F4/80(+)
cells and the activation of the p38 MAPK-NF-κB pathway in CD4(+) T cells and
macrophages [156, 165]. Emodin was capable of improving the lipid accumulation
through the ERS-SREBP1c pathway in fructose-induced nonalcoholic fatty liver
disease [75]. Thus emodin might be applied as a potential candidate for the pre-
vention and intervention of liver diseases. However, in a recent study toxic effects
of emodin were reported in idiosyncratic liver injury model by Tu et al. [132].
Emodin-potentiated liver injury induced by noninjurious dose of LPS. The mech-
anisms underlying this effect are yet to be fully understood.
Emodin has been reported to have anti-inflammatory effects in both in vitro and
in vivo system (Tables 3.1, 3.2). Emodin when administered intraperitoneally
reduced LPS-induced mammary gland injury in mastitis [76]. Emodin has also
proved to be a potential candidate in treating serious vascular inflammatory dis-
eases, such as sepsis and septic shock and had anti-inflammatory effects in sepsis
mouse model in vivo [64, 65]. Emodin inhibited inflammation in intestinal
epithelial cells by blocking HIF-1α/NF-κB-COX-2 signaling pathways [67].
Emodin also lessened ocular tissue inflammation and fibrosis by inhibition of
NF-κB pathway after eye injury [11, 55]. Lee et al. [66] reported anti-inflammatory
effects of emodin derivative [6-O-β-D-glucoside emodin (EG)] in vitro in human
umbilical vein endothelial cells (HUVECs) as well as in mice. Recently, Han et al.
[32] reported that emodin attenuated NLRP3 inflammasome activation, leading to
decreased secretion of cleaved IL-1β, and blocking of the inflammasome-induced
pyroptosis. Ocular neovascularization is the origin of blindness related with
ischemic retinal ataxia in conjunction with proliferative diabetic retinopathy (PDR),
retinopathy of prematurity (ROP) and age-related macular degeneration. The
induction of angiogenesis in retina is due to VEGF. Emodin-MgSiO3 nanoparticles
could inhibit the expression of both VEGF gene and protein effectively and can be
an effective therapy for eye-related disorders [106].
PI3K
Inflammosome MAPK
NF-kβ Akt
ERK JNK P38
mTOR GSK3
IL-1β
References
10. Chen C, Liang Z, Chen Q, Li ZG (2012) Irbesartan and emodin on myocardial remodelling in
Gold Blatt hypertensive rats. J Cardiovasc Pharmacol 60(4):375–380
11. Chen G, Zhang J, Zhang H, Xiao Y, Kao X, Liu Y, Liu Z (2015) Anti-Inflammatory effect
of emodin on lipopolysaccharide-induced keratitis in wistar rats. Int J ClinExp Med
8(8):12382–12389
12. Chen Q, Pang L, Huang S, Lei W, Huang D (2014) Affects of emodin and irbesartan on
ventricular fibrosis in Gold Blatt hypertensive rats. Pharmacies 69(5):374–378
13. Chen YC, Shen SC, Lee WR, Hsu FL, Lin HY, Ko CH, Tseng SW (2002) Emodin induces
apoptosis in human promyeloleukemic HL-60 cells accompanied by activation of caspase 3
cascades but independent of reactive oxygen species production. Biochem Pharmacol
64:1713–1721
14. Chen Z, Zhang L, Yi J, Li Z (2012) Promotion of adiponectin multimerization by emodin: A
novel AMPK activator with PPARγ-agonist activity. J Cell Biochem 113(11):3547–3558
15. Chien SC, Wu YC, Chen ZW, Yang WC (2015) Naturally occurring anthraquinones:
chemistry and therapeutic potential in autoimmune diabetes. Evid Based Complem Altern
Med, Article ID 357357
16. Chihara T, Shimpo K, Beppu H, Yamamoto N, Kaneko T, Wakamatsu K, Sonoda S (2015)
Effects of aloe-emodin and emodin on proliferation of the MKN45 human gastric cancer cell
line. Asian Pac J Cancer Prev 16(9):3887–3891
17. Chu X, Wei M, Yang X, Cao Q, Xie X, Guan M, Wang D, Deng X (2012) Effects of an
anthraquinone derivative from Rheum Officinale Baill, Emodin, on airway responses in a
murine model of asthma. Food Chem Toxicol 50(7):2368–2375
18. Dang SS, Zhang X, Jia XL, Cheng YA, Song P, Liu EQ, He Q, Li ZF (2008) Protective
effects of emodin and astragalus polysaccharides on chronic hepatic injury in rats. Chin
Med J (Engl) 121(11):1010–1014
19. Deng G, Ju X, Meng Q, Yu ZJ, Ma LB (2015) Emodin inhibits the proliferation of PC3
prostate cancer cells in vitro via the notch signalling pathway. Mol Med Rep 12(3):
4427–4433
20. Dong H, Lu FE, Gao ZQ, Xu LJ, Wang KF, Zou X (2005) Effects of emodin on treating
murine nonalcoholic fatty liver induced by high caloric laboratory chaw. World J
Gastroenterol 11(9):1339–1344
21. Dong H, Lu FE, Gao ZQ (2006) Experimental Study on effects of emodin on nonalcohoalic
fatty liver induced by high fat diet in rats. Zhongguo Zhong Xi Yi Jie He Za Zhi 26 suppl:
64–67
22. Dong MX, Jia Y, Zhang YB, Li CC, Geng YT, Zhou L, Li XY, Liu JC, Niu YC (2009)
Emodin protects rat liver from CCl(4)-induced fibrogenesis via inhibition of hepatic stellate
cells activation. World J Gastroenterol 15(38):4753–4762
23. Du Y, Ko KM (2006) Effects of pharmacological preconditioning by emodin/oleanolic acid
treatment and/or ischemic preconditioning on mitochondrial antioxidant components as well as
the susceptibility to ischemia-reperfusion injury in rat hearts. Mol Cell Biochem 288(1–2):
135–142
24. Evans WC (1996) Treas and evanspharmacognosy, 4th edn. W.B. Saunders Company,
London
25. Fang SH, Hou YC, Chang WC, Hsiu SL, Chao PD, Chiang BL (2003) Morin sulphates/
glucuronides exert anti-inflammatory activity on activated macrophages and decreased the
incidence of septic shock. Life Sci 743–756
26. Feng Y, Huang SL, Dou W, Zhang S, Chen JH, Shen Y, Shen JH, Leng Y (2010) Emodin, a
natural product, selectively inhibits 11b-hydroxysteroid dehydrogenase type 1 and
ameliorates metabolic disorder in diet-induced obese mice. Br J Pharmacol 161:113–126
27. Fu JM, Zhou J, Shi J, Xie JS, Huang L, Yip AY, Loo WT, Chow LW, Ng EL (2012) Emodin
affects ERCC1 expression in breast cancer cells. J Transl Med 19(10 Suppl 1):S7
28. Gao J, Wang F, Wang W, Su Z, Guo C, Cao S (2014) Emodin suppresses hyperglycaemia-
induced proliferation and fibronectin expression in mesangial cells via inhibiting
cflip. PLoS ONE 9(4):e93588
66 B.A. Monisha et al.
29. Gao Y, Liu H, Deng L, Zhu G, Xu C, Li G, Liu S, Xie J, Liu J, Kong F, Wu R, Li G, Liang S
(2011) Effect Of emodin on neuropathic pain transmission mediated by P2X2/3 receptor of
primary sensory neurons. Brain Res Bull 84(6):406–413
30. Gao Y, Zhang J, Li G, Xu H, Yi Y, Wu Q, Song M, Bee YM, Huang L, Tan M, Liang S,
Li G (2015) Protection of vascular endothelial cells from high glucose-induced cytotoxicity
by emodin. Biochem Pharmacol 94(1):39–45
31. Ha MK, Song YH, Jeong SJ, Lee HJ, Jung JH, Kim B, Song HS, Huh JE, Kim SH (2011)
Emodin inhibits proinflammatory responses and inactivates histone deacetylase 1 in hypoxic
rheumatoid synoviocytes. Biol Pharm Bull 34(9):1432–1437
32. Han JW, Shim DW, Shin WY, Heo KH, Kwak SB, Sim EJ, Jeong JH, Kang TB, Lee KH
(2015) Anti-inflammatory effect of emodin via attenuation of NLRP3 inflammasome
activation. Int J Mol Sci 16:8102–8109
33. Harborne JB, Baxter H, Moss GP (1999) Phytochemical dictionary. A handbook of bioactive
compounds from plants, 2nd edn. Taylor & Francis, London
34. He L, Bi JJ, Guo Q, Yu Y, Ye XF (2012) Effects of emodin extracted from chinese herbs on
proliferation of non-small cell lung cancer and underlying mechanisms. Asian Pac J Cancer
Prev 13(4):1505–1510
35. He Z, He MF, Ma SC, But PP (2009) Anti-angiogenic effects of rhubarb and its
anthroquinone derivatives. J Ethnopharmacol 121313–121317
36. Heo SK, Yun HJ, Park WH, Park SD (2008) Emodin inhibits TNF-Α-induced human aortic
smooth-muscle cell proliferation via caspase and mitochondrial-dependent apoptosis. J Cell
Biochem 105(1):70–80
37. Hsu CM, Hsu YA, Tsai Y (2010) Emodin inhibits the growth of hepatoma cells: finding the
common anti-cancer pathway using Huh7, Hep3B, Andhepg2 cells. Biochem Biophys Res
Commun 392(4):473–478
38. Hu H, Sun W, Gu LB, Tu Y, Liu H (2015) Molecular mechanism of emodin on inhibiting
autophagy induced by HBSS in renal tubular cells. Zhongguo Zhong Yao ZaZhi 40(10):
1965–1970
39. Huang HC, Chu SH, Chao-Lee PD (1991) Vasorelaxants from Chinese herbs, emodin and
scoparone, possess immunosuppressive properties. Eur J Pharmacol 198:211–213
40. Huang LY, Hu JD, Chen XJ, Zhu LF, Hu HL (2005) Effects of emodin on the proliferation
inhibition and apoptosis induction in HL-60 cells and the involvement of c-myc gene.
Zhonghua Xue Ye XueZaZhi 26(6):348–351
41. Hwang JK, Noh EM, Moon SJ, Kim JM, Kwon KB, Park BH (2013) Emodin suppresses
inflammatory responses and joint destruction in collagen-induced arthritic mice.
Rheumatology (Oxf Engl) 52(9):1583–1591
42. Hwang SY, Heo K, Kim JS, Im JW, Lee SM, Cho M, Kang DH, Heo J, Lee JW, Choi CW,
Yang K (2015) Emodin attenuates radio-resistance induced by hypoxia in HepG2 cells
via the enhancement of PARP1 cleavage and inhibition of JMJD2B. Oncol Rep 33(4):
1691–1698
43. Izhaki I (2002) Emodin—a secondary metabolite with multiple ecological functions in higher
plants. New Phytol 155:205–217
44. Jelassi B, Anchelin M, Chamouton J, Cayuela ML, Clarysse L, Li J, Gore J, Jiang LH,
Roger S (2013) Anthraquinone emodin inhibits human cancer cell invasiveness by
antagonizing P2X7 receptors. Carcinogenesis 34(7):1487–1496
45. Jia X, Fang Yu, Wang J, Iwanowycz S, Saaoud F, Wang Y, Hu J, Wang Q, Fan D (2014)
Emodin suppresses pulmonary metastasis of breast cancer cells accompanied with decreased
macrophage recruitment and M2 polarization in the lungs. Breast Cancer Res Treat 148
(2):291–302
46. Jiang N, Liao W, Kuang X (2014) Effects of emodin on IL-23/IL-17 inflammatory axis, Th17
cells and viral replication in mice with viral myocarditis. J South Med Univ 34(3):373–378
47. Jin JH, Ngoc TM, Bae KH, Kim YS, Kim HP (2011) Inhibition of experimental atopic
dermatitis by rhubarb (rhizomes of rheum tanguticum) and 5-lipoxygenase inhibition of its
major constituent. Emodin Phytother Res 25(5):755–775
3 Emodin and Its Role in Chronic Diseases 67
48. Jing X, Ueki N, Cheng J, Imanishi H, Hada T (2002) Induction of apoptosis in hepatocellular
carcinoma cell lines by Emodin. Jpn J Cancer Res 93:874–882
49. Jun C, Niu X, Chen Y, Hu Q, Shi G, Wu H, Wang J, Yi J (2008) Emodin-induced generation
of reactive oxygen species inhibits RhoA activation to sensitize gastric carcinoma cells to
anoikis. Neoplasia 10:41–51
50. Kamei H, Koide T, Kojima T, Hashimoto Y, Hasegawa M (1998) Inhibition of cell growth in
culture by quinones. Cancer Biother Radiopharm 13(3):185–188
51. Kaneshiro T, Morioka T, Inamine M, Kinjo T, Arakaki J, Chiba I, Sunagawa N, Suzui M,
Yoshimi N (2006) Anthraquinone derivative emodin inhibits tumor-associated angiogenesis
through inhibition of extracellular signal-regulated kinase 1/2 phosphorylation. Eur J
Pharmacol 553:46–53
52. Kang DM, Yoon KH, Kim JY, Oh JM, Lee M, Jung ST, Juhng SK, Lee YH (2014) CT
imaging bio-marker for evaluation of emodin as a potential drug on LPS-mediated
osteoporosis mice. Acad Radiol 21(4):457–462
53. Kim DY, Kang TB, Shim DW, Sun X, Han JW, Ji YE, Kim TJ, Koppula S, Lee KH (2014)
Emodin attenuates A23187-induced mast cell degranulation and tumor necrosis factor-Α
secretion through protein kinase C and Iκb kinase 2 signaling. Eur J Pharmacol 723:501–506
54. Kim JY, Cheon YH, Kwak SC, Baek JM, Yoon KH, Su M, Oh LJ (2014) Emodin regulates
bone remodelling by inhibiting osteoclastogenesis and stimulating osteoblast formation.
J Bone Miner Res 29(7):1541–1553
55. Kitano AI, Saika S, Yamanaka O, Ikeda K, Okada Y, Shirai K, Reinach PS (2007) Emodin
suppression of ocular surface inflammatory reaction. Invest Ophthalmol Vis Sci 489
(11):5013–5022
56. Kogl F, Postowsky JJ (1925) Untersuchungen üuber Pilzfarbstoffe. II. Uber die Farbstaffe des
blutroten Hautkorpfes (Dermocybe sanquinea Wulf.). Justus LiebigsAnnalen der Chemie
444:1–7
57. Koyama M, Kelly TR, Watanabe KA (1988) Novel type of potential anticancer agents
derived from chrysophanol and emodin. J Med Chem 31:283–284
58. Kuo TC, Yang JS, Lin MW, Hsu SC, Lin JJ, Lin HJ (2009) Emodin has cytotoxic and
protective effects in rat C6 glioma cells: roles of Mdr1a and nuclear factor B in cell survival.
J Pharmacol Exp Ther 330(3):736–744
59. Kuo YC, Sun CM, Ou JC, Tsai WJ (1997) A tumor cell growth inhibitor from polygonum
hypoleucum ohwi. Life Sci 61(23):2335–2344
60. Kwak HJ, Park MJ, Park CM, Moon SI, Yoo DH, Lee HC, Lee SH, Kim MS, Lee HW,
Shin WS, Park IC, Rhee CH, Si Hong (2006) Emodin inhibits vascular endothelial growth
factor-A-induced angiogenesis by blocking receptor-2 (KDR/Flk-1) phosphorylation. Int J
Cancer 118:2711–2720
61. Lee BH, Huang YY, Duh PD, Wu SC (2012) Hepatoprotection of emodin and polygonum
multiflorum against Ccl(4)-induced liver injury. Pharm Biol 50(3):351–359
62. Lee HZ (2001) Protein kinase C involvement in aloe-emodin- and emodin-induced apoptosis
in lung carcinoma cell. Br J Pharmacol 134:1093–1102
63. Lee J, Jung E, Lee J, Huh S, Hwang CH, Lee HY, Kim EJ, Cheon JM, Hyun CG, Kim YS,
Park D (2006) Emodin inhibits TNF alpha-induced MMP-1 expression through suppression
of activator protein-1 (AP-1). Life Sci 79:2480–2485
64. Lee W, Ku SK, Kim TH, Bae JS (2013) Emodin-6-O-B-D-glucoside inhibits HMGB1-
induced inflammatory responses in vitro and in vivo. Food Chem Toxicol 52:97–104
65. Lee W, Ku SK, Bae JS (2013) Emodin-6-O-Β-D-glucoside down-regulates endothelial
protein C receptor shedding. Arch Pharm Res 36(9):1160–1165
66. Lee W, Ku SK, Lee D, Lee T, Bae JS (2014) Emodin-6-O-Β-D-glucoside inhibits
high-glucose-induced vascular inflammation. Inflammation 37(2):306–313
67. Lei Q, Qiang F, Chao D, Di W, Guoqian Z, Bo Y, Lina Y (2014) Amelioration of hypoxia
and LPS-induced intestinal epithelial barrier dysfunction by emodin through the suppression
of the NF-Κb and HIF-1α signaling pathways. Int J Mol Med 34(6):1629–1639
68 B.A. Monisha et al.
87. Ljubimov AV, Caballero S, Agoki AM, Pinna LA, Grant MB (2004) Involvement of protein
kinase CK2 in angiogenesis and retinal neovascularization. Invest Ophthalmol Vis Sci
45:4583–4591
88. Lu Y, Yang JH, Li X, Hwangbo K, Hwang SL, Taketomi Y, Murakami M, Chang YC,
Kim CH, Son JK, Chang HW (2011) Emodin, a naturally occurring anthroquinone
derivative, suppresses IGE-mediated anaphylactic reaction and mast cell activation. Biochem
Pharmacol 82(11):1700–1708
89. Ma J, Lu H, Wang S, Chen B, Liu Z, Ke X, Liu T, Fu J (2015) the anthraquinone derivative
emodin inhibits angiogenesis and metastasis through downregulating Runx2 activity in breast
cancer. Int J Oncol 46(4):1619–1628
90. Ma J, Yang J, Wang C, Zhang N, Dong Y, Wang C, Wang Y, Lin X (2014) Emodin
augments cisplatin cytotoxicity in platinum-resistant ovarian cancer cells via ros-dependent
MRP1 downregulation. Biomed Res Int: Article ID 107671, 8 p
91. Ma Y-S, Weng SW, Lin MW, Lu CC, Chiang JH, Yang JS, Lai KC, Lin JP, Tang NY,
Lin JG, Chung JG (2012) Antitumor effects of emodin on LS1034 human colon cancer cells
in vitro and in vivo: roles of apoptotic cell death and LS1034 tumor xenografts model. Food
Chem Toxicol 50:1271–1278
92. Markovic ZS, Manojlovic NT (2009) DFT study on the reactivity of OH groups in emodin.
Monatsh Chem 140:1311–1318
93. Masaldan S, Iyer VV (2014) Exploration of effects of emodin in selected cancer cell lines:
enhanced growth inhibition by ascorbic acid and regulation of LRP1 and AR under
hypoxia-like conditions. J Appl Toxicol 34(1):95–104
94. Meng KW, Lv Y, Yu L, Wu SL, Pan CE (2005) Effects of emodin and double blood supplies
on liver regeneration of reduced size graft liver in rat model. World J Gastroenterol 11
(19):2941–2944
95. Meng L, Yan D, Xu W, Ma J, Chen B, Feng H (2012) Emodin inhibits tumor necrosis
factor-α-induced migration and inflammatory responses in rat aortic smooth muscle cells.
Int J Mol Med 29(6):999–1006
96. Mizuno M, Kawamura H, Takei N, Nawa H (2008) The anthraquinone derivative emodin
ameliorates neurobehavioral deficits of a rodent model for schizophrenia. J Neural Transm
(Vienna) 115(3):521–530
97. Mueller SO, Schmitt M, Dekant W, Stopper H, Schlatter J, Schreier P, Lutz WK (1999)
Occurrence of emodin, chrysophanol and physcion in vegetables, herbs and liquors.
Genotoxicity and anti-genotoxicity of the anthraquinones and of the whole plants. Food
Chem Toxicol 37:481–491
98. Muto A, Hori M, Sasaki Y, Saitoh A, Yasuda I, Maekawa T, Uchida T, Asakura K,
Nakazato T, Kaneda T, Kizaki M, Ikeda Y, Yoshida T (2007) Emodin has a cytotoxic
activity against human multiple myeloma as a janus-activated kinase 2 inhibitor. Mol Cancer
Ther 6(3):987–994
99. Nemmar A, Al-Salam S, Yuvaraju S, Beegam S, Ali BH (2015) Emodin mitigates diesel
exhaust particles-induced increase in airway resistance, inflammation and oxidative stress in
mice. Respir Physiol Neurobiol 15(215):51–57
100. Ni Q, Sun K, Chen G, Shang D (2014) In vitro effects of emodin on peritoneal macrophages
that express membrane-bound CD14 protein in a rat model of severe acute
pancreatitis/systemic inflammatory response syndrome. Mol Med Rep 9:355–359
101. Ni Q, Zhang W, Sun K, Yin C, An J, Shang D (2014) In vitro effects of emodin on peritoneal
macrophage intercellular adhesion molecule-3 in a rat model of severe acute
pancreatitis/systemic inflammatory response syndrome. Biomed Rep 2(1):63–68
102. Ok S, Kim SM, Kim C, Nam D, Shim BS, Kim SH, Ahn KS, Choi SH, Ahn KS (2012)
Emodin inhibits invasion and migration of prostate and lung cancer cells by downregulating
the expression of chemokine receptor CXCR4. Immunopharmacol Immunotoxicol 34(5):
768–778
70 B.A. Monisha et al.
122. Subramaniam A, Shanmugam MK, Ong TH, Li F, Perumal E, Chen L, Vali S, Abbasi T,
Kapoor S, Ahn KS, Kumar AP, Hui KM, Sethi G (2013) Emodin inhibits growth and induces
apoptosis in an orthotopic hepatocellular carcinoma model by blocking activation of STAT3.
Br J Pharmacol 170(4):807–821
123. Sui JQ, Xie KP, Zou W, Xie MJ (2014) Emodin inhibits breast cancer cell proliferation
through the Erα-MAPK/Akt-Cyclin D1/Bcl-2 signaling pathway. Asian Pac J Cancer Prev 15
(15):6247–6251
124. Sun Y, Wang X, Zhou Q, Lu Y, Zhang H, Chen Q, Zhao M, Su S (2015) Inhibitory effect of
emodin on migration, invasion and metastasis of human breast cancer MDA-MB-231 cells
in vitro and in vivo. Oncol Rep 33(1):338–346
125. Sun Y, Sun L, Liu S, Song J, Cheng J, Liu J (2015) Effect of emodin on aquaporin 5
expression in rats with sepsis-induced acute lung injury. J Tradit Chin Med 35(6):679–684
126. Sun YP, Liu JP (2015) Blockade of emodin on amyloid-Β 25-35-induced neurotoxicity in
Aβpp/PS1 mice and PC12 cells through activation of the class III phosphatidylinositol
3-Kinase/Beclin-1/B-cell lymphoma 2 pathway. Planta Med 81(2):108–115
127. Sun ZH, Bu P (2012) Downregulation of phosphatase of regenerating liver-3 is involved in
the inhibition of proliferation and apoptosis induced by emodin in the SGC-7901 human
gastric carcinoma cell line. Exp Ther Med 3(6):1077–1081
128. Tang Q, Zhao S, Wu J, Zheng F, Yang L, Hu J, Hann SS (2015) Inhibition of integrin-linked
kinase expression by emodin through crosstalk of Ampkα and ERK1/2 signaling and
reciprocal interplay of Sp1 and C-Jun. Cell Signal 27:1469–1477
129. Thacker PC, Karunagaran D (2014) Emodin suppresses Wnt signaling in human colorectal
cancer cells SW480 and SW620. Eur J Pharmacol 5(742):55–64
130. Thacker PC, Karunagaran D (2015) Curcumin and emodin down-regulate TGF-β signaling
pathway in human cervical cancer cells. PLoS ONE 10(3):e0120045
131. Thomson RH (1987) Naturally occurring quinines. III. Recent advances. London
132. Tu C, Gao D, Li XF, Li CY, Li RS, Zhao YL, Li N, Jia GL, Pang JY, Cui HR, Ma ZJ,
Xiao XH, Wang JB (2015) Inflammatory stress potentiates emodin-induced liver injury in
rats. Front Pharmacol 23(6):233
133. Tzeng TF, Lu HJ, Liou SS, Chang CJ, Liu IM (2012) Emodin, a naturally occurring
anthraquinone derivative, ameliorates dyslipidemia by activating AMP-activated protein
kinase in high-fat-diet-fed rats. Evid Based Complement Altern Med 2012:781812
134. Van den Berg AJJ, Labadie RP (1989) Quinones. In: Deypm H (ed) Methods in plant
biochemistry, vol 1. Plant phenolics. Academic Press, London, pp 451–491
135. van Gorkom BA, De Vries EG, Karrenbeld A, Kleibeuker JH (1999) Anthranoid laxatives
and their potential carcinogenic effects. Aliment Pharmacol Ther 13(4):443–452
136. Waly MI, Ali BH, Al-Lawati I, Nemmar A (2013) Protective effects of emodin against
cisplatin-induced oxidative stress in cultured human kidney (HEK 293) cells. J Appl Toxicol
33:626–630
137. Wang J, Huang H, Liu P, Tang F, Qin J, Huang W, Chen F, Guo F, Liu W, Yang B (2006)
Inhibition of phosphorylation of P38 MAPK involved in the protection of nephropathy by
emodin in diabetic rats. Eur J Pharmacol 553(1–3):297–303
138. Wang RT, Yin H, Dong SB, Yuan W, Liu YP, Liu C (2014) Research progress of emodin
anti-gallbladder carcinoma. Zhongguo Zhong Yao Za Zhi Zhongguo Zhong Yao Za Zhi 39
(11):1976–1978
139. Wang T, Zhong XG, Li YH, Jia X, Zhang SJ, Gao YS, Liu M, Wu RH (2015) Protective
effect of emodin against airway inflammation in the ovalbumin-induced mouse model. Chin J
Integr Med. 21(6):431–437
140. Wang W, Sun Y, Li X, Li H, Chen Y, Tian Y, Yi J, Wang J (2011) Emodin potentiates the
anticancer effect of cisplatin on gallbladder cancer cells through the generation of reactive
oxygen species and the inhibition of survivin expression. Oncol Rep 26(5):1143–1148
141. Wang W, Sun YP, Huang XZ, He M, Chen YY, Shi GY, Li H, Yi J, Wang J (2010) Emodin
enhances sensitivity of gallbladder cancer cells to platinum drugs via glutathion depletion
and MRP1 downregulation. Biochem Pharmacol 79(8):1134–1140
72 B.A. Monisha et al.
142. Wang W, Zhou Q, Liu L, Zou K (2012) Anti-allergic activity of emodin on IgE-mediated
activation in RBL-2H3 cells. Pharmacol Rep 64(5):1216–1222
143. Wang XH, Wu SY, Zhen YS (2004) Inhibitory effects of emodin on angiogenesis. Yao Xue
Xue Bao 39(4):254–258
144. Wang XJ, Yang J, Cang H, Zou YQ, Yi J (2005) Gene expression alteration during redox
dependent enhancement of arsenic cytotoxicity by emodin in hela cells. Cell Res 15(7):
511–522
145. Wang YJ, Huang SL, Feng Y, Ning MM, Leng Y (2012) Emodin, an 11β-hydroxysteroid
dehydrogenase type 1 inhibitor, regulates adipocyte function in vitro and exerts anti-diabetic
effect in Ob/Ob mice. Acta Pharmacol Sin 33(9):1195–1203
146. Way TD, Husang JT, Chou CH, Huang CH, Yang MH, Ho CT (2014) Emodin represses
tWIST 1-induced epithelial-mesenchymal transitions in head and neck squamous cell
carcinoma cells by inhibiting the β-catenin and Akt pathways. Eur J Cancer 50:366–378
147. Wei WT, Chen H, Ni ZL, Liu HB, Tong HF, Fan L, Liu A, Qiu MX, Liu DL, Guo HC,
Wang ZH, Lin SZ (2011) Antitumor and apoptosis-promoting properties of emodin, an
anthraquinone derivative from rheum officinale baill, against pancreatic cancer in mice via
inhibition of Akt activation. Int J Oncol 39(6):1381–1390
148. Wu W, Hu N, Zhang Q, Li Y, Li P, Yan R (2014) In vitro glucuronidation of five rhubarb
anthraquinones by intestinal and liver microsomes from humans and rats. Chem Biol Interact
5(219):18–27
149. Wu Y, Tu X, Lin G, Xia H, Huang H, Wan J, Cheng Z, Liu M, Chen G, Zhang H, Fu J,
Liu Q, Liu DX (2007) Emodin-mediated protection from acute myocardial infarction via
inhibition of inflammation and apoptosis in local ischemic myocardium. Life Sci 81(17–18):
1332–1338
150. Wu Z, Chen Q, Ke D, Li G, Deng W (2014) Emodin protects against diabetic
cardiomyopathy by regulating the AKT/GSK-3β signaling pathway in the rat model.
Molecules 19(9):14782–14793
151. Wu ZX, Yu BP, Xu L (2009) Emodin inhibits voltage-dependent potassium current in guinea
pig gallbladder smooth muscle. Basic Clin Pharmacol Toxicol 105(3):167–172
152. Xiang MX, Xu Z, Su HW, Hu J, Yan YJ (2011) Emodin-8-O-Β-D-glucoside from
polygonum amplexicaule D. Don Var. Sinense Forb promotes proliferation and
differentiation of osteoblastic MC3T3-E1 cells. Molecules 16(1):728–737
153. Xiao M, Zhu T, Zhang W, Wang T, Shen YC, Wan QF, Wen FQ (2014) Emodin ameliorates
LPS-induced acute lung injury, involving the inactivation of NF-κB in mice. Int J Mol Sci 15
(11):19355–19368
154. Xing JY, Song GP, Deng JP, Jiang LZ, Xiong P, Yang BJ, Liu SS (2015) Antitumor effects
and mechanism of novel emodin rhamnoside derivatives against human cancer cells in vitro.
PLoS One 10(12):e0144781
155. Xue J, Ding W, Liu Y (2010) Anti-diabetic effects of emodin involved in the activation of
PPARγ on high-fat diet-fed and low dose of streptozotocin-induced diabetic mice. Fitoterapia
81(3):173–177
156. Xue J, Chen F, Wang J, Wu S, Zheng M, Zhu H, Liu Y, He J, Chen Z (2015) Emodin
protects against concanavalin a-induced hepatitis in mice through inhibiting activation of the
p38 MAPK-NF- κB signaling pathway. Cell Physiol Biochem 35(4):1557–1570
157. Xue WH, Shi XQ, Liang SH, Zhou L, Liu KF, Zhao J (2015) Emodin attenuates cigarette
smoke induced lung injury in a mouse model via suppression of reactive oxygen species
production. J Biochem Mol Toxicol 29(11):526–532
158. Yan YY, Zheng LS, Zhang X, Chen LK, Singh S, Wang F, Zhang JY, Liang YJ, Dai CL,
Gu LQ, Zeng MS, Talele TT, Chen ZS, Fu LW (2011) Blockade of Her2/neu binding to
Hsp90 by emodin azide methyl anthraquinone derivative induces proteasomal degradation of
Her2/neu. Mol Pharm 8(5):1687–1697
159. Yang JH, Hsia TC, Kuo HM, Chao PD, Chou CC, Wei YH, Chung JG (2006) Inhibition of
lung cancer cell growth by quercetin glucuronides via G2/M arrest and induction of
apoptosis. Drug Metab Dispos 34(2):296–304
3 Emodin and Its Role in Chronic Diseases 73
Abstract Chronic diseases pose a worldwide problem and are only continuing to
increase in incidence. Two major factors contributing to the increased incidence in
chronic disease are a lack of physical activity and poor diet. As the link between
diet and lifestyle and the increased incidence of chronic disease has been well
established in the literature, novel preventive, and therapeutic methods should be
aimed at naturally derived compounds such as ursolic acid (UA), the focus of this
chapter. As chronic diseases, obesity and cancer share the common thread of
inflammation and dysregulation of many related pathways, the focus here will be on
these two chronic diseases. Significant evidence in the literature supports an
important role for natural compounds such as UA in the prevention and treatment of
chronic diseases like obesity and cancer, and here we have highlighted many of the
ways UA has been shown to be a beneficial and versatile phytochemical.
4.1 Introduction
Over the past 50 years, trends in lifestyle and diet have drastically affected the way
human beings experience disease and aging. Tragically, the modern western diet is
poorly balanced and composed of many nutrient-deficient foods. With this in mind,
A.M. Mancha-Ramirez
Department of Cellular and Structural Biology, The University of Texas Health
Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
e-mail: [email protected]
T.J. Slaga (&)
Department of Pharmacology, The University of Texas Health Science Center
San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
e-mail: [email protected]
it is not surprising that chronic disease poses a serious worldwide problem, and one
that has undoubtedly risen from a stark dissonance between today’s modern diet
and that which our ancestors once thrived upon.
The modern western diet is characterized by an excess of salt and refined sugar,
and the readily available low cost, high calorie, nutrient foods that are part of a
typical western diet are believed to be a key component in the increasing obesity
epidemic [77, 116]. Specifically, based on data obtained from the National Health
and Nutrition Examination Survey, some of the most concerning issues with the
typical modern western diet include an increased uptake of refined grains, added
sugars, and saturated fats [81]. Moreover, it has been suggested that some of the
modern agricultural practices and over-processed nature of the typical western diet
has contributed to the nutrient deficiency seen in many components of regularly
consumed food products [116].
As the link between diet and lifestyle and the increased incidence of chronic
disease has been well established in the literature, it makes sense to direct our
attention to phytonutrients as meaningful therapeutic agents for the prevention and
treatment of chronic illnesses such as those discussed in this chapter. Natural
remedies derived from plants play an important role both in the prevention of
chronic disease development and in the treatment of chronic inflammation-driven
diseases. Moreover, plant-derived compounds have been the primary source of
medication for centuries in the Asian subcontinent [3, 7].
Significant evidence in the literature suggests that persistent inflammation is the
primary initiating factor that causes major chronic disease. Unfortunately, novel
approaches towards prevention or treatment of chronic diseases is still moving quite
slowly [1]. It is well known that phytonutrients derived from plants and their
extracts have been widely used in Eastern medicine for thousands of years for a
variety of illnesses included but not limited to hypertension, inflammation, and of
course cancer [58, 74]. Modern epidemiological studies have shown that con-
sumption of nutrient-rich fruit and vegetable-based diets can decrease the risk of
many diseases including metabolic syndrome and cancer [10, 43, 70, 128]. One
such powerful and versatile phytonutrient is ursolic acid (UA), a pentacyclic
triterpene that can be found in apples and rosemary, among other sources [40]. In
this chapter, we will discuss this phytonutrient in great detail including the multi-
tude of ways UA has shown great efficacy in both metabolic disorder maladies as
well as against several types of cancer (Fig. 4.1).
When considering the importance of chronic illness, for perspectives sake, let us
picture a crowded football stadium where every seat is filled; now, imagine that the
people in this stadium represent the population of the United States. According to a
2012 CDC report, approximately half of the fans in our imaginary stadium had one
or more chronic health condition(s), and one in four had two or more chronic health
conditions. Stepping away from our stadium analogy, this translates to approxi-
mately 117 million Americans, half of the adult population, who were living with
chronic illnesses in 2012. Of the ten most common chronic diseases, this chapter will
focus on cancer, obesity, and other illnesses that result from the latter, such as those
associated with metabolic syndrome. Perhaps after using our imagination for the
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 77
football stadium exercise, it is not difficult to see why chronic diseases and condi-
tions are among the costliest of all health problems. However, what is difficult to
discern is why we have not managed to diminish this increasing trend of chronic
illness when much of it can be prevented. The unfortunate trend in poor diet choices
coupled with incredibly busy lifestyles, which do not leave any room for regular
exercise, has led to a startling incidence in obesity and subsequently, the rise of
metabolic syndrome in the western world. Metabolic syndrome is a generalized term
for maladies such as hypertension, heart disease, and type II diabetes, all of which
are typically developed as a consequence of an individual having low physical
activity, resulting in obesity. Interestingly, there are several different types of cancer
including colon and breast cancer where risk factor is greatly increased by being
overweight or obese. Consequently, there is growing evidence supporting a strong
link between metabolic syndrome and an increased risk of developing cancer,
making it quite easy to see how these two chronic illnesses are tightly intertwined.
According to the World Health Organization, being overweight is one of the top 10
risk conditions in the world, and one which will affect upwards of one billion people
by 2030 [37]. Perhaps the most devastating aspect of this chronic illness is the fact
that obesity brings about so many different comorbidities that can significantly
compromise a patient’s quality of life; one highly relevant example of this is dia-
betes. Current predictions for the prevalence of diabetes suggest that this disease will
globally increase from 285 million in 2010 to 439 million in 2030 [101]. Not only
has diabetes quickly become an epidemic, but the other characteristic aspects of the
disease such as damage to tissue in the liver, kidney, adipose tissue, pancreas, and
vasculature as a result of pro-inflammation and oxidative stress, generate unique
problems of their own [14]. It is vital to remember that the pathogenesis of obesity is
extremely complex and can be caused by a variety of factors such as a genetic
predisposition, metabolism, physiology, endocrine problems, and behavioral issues
78 A.M. Mancha-Ramirez and T.J. Slaga
Although cancer and obesity are two entirely different diseases by definition, they
share many cellular signaling pathways that play a part in their pathogenesis.
Moreover, both cancer and obesity-related diseases share a very important aspect of
pathogenesis, which is inflammation. Chronic inflammation has been linked with
most chronic illnesses including cancer, cardiovascular disease, diabetes, obesity,
and neurologic disease [3]. Significant epidemiological evidence shows an increased
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 79
risk of acquiring several different types of cancer which is greatly increased by being
overweight or obese [5]. Specifically, a wide variety of cancers, including high-
grade prostate cancer, colorectal cancer, postmenopausal breast cancer, and mela-
noma have been shown to be linked to obesity in epidemiological studies [15, 25, 92,
105]. As you might expect, just as obesity and diabetes have been shown to be
associated with increase cancer incidence, conversely, exercise, and decreased
caloric intake have been shown to be associated with decreased cancer incidence. It
is well known that calorie restriction and exercise aid in the prevention of obesity
and diabetes, but both have also been correlated with decreased cancer incidences in
many epidemiological studies and can directly inhibit tumor formation in certain
cancer models [48, 75, 79]. For example, calorie restriction was able to prevent
tumor formation in various mouse models of cancer including the breast, colon,
brain, prostate, and chemically induced skin cancer [24, 69, 102].
When there is excess dietary intake in an individual, the result is excess fat
storage and improper processing of glucose and lipids [32]. The peptide hormone
insulin is secreted by the b cells of the pancreas and its job is to maintain normal
blood glucose levels by facilitating cellular glucose uptake, regulating carbohy-
drate, lipid, and protein metabolism and promoting cell division and growth [122].
The goal of insulin in healthy humans is to increase glucose uptake and tell the liver
to shut down the process of gluconeogenesis. Conversely, in overweight or obese
humans, this process is derailed by chronic exposure to high levels of glucose and
free fatty acids. Obesity-associated inflammation regulates insulin resistance in
target cells, ultimately resulting in more insulin production in order to reduce blood
glucose and fatty acid levels [51, 122]. The end result of the increased demand for
insulin production is the breakdown of insulin-producing b-cells in the pancreas,
and ultimately type II diabetes (Fig. 4.2).
Fig. 4.2 The role of obesity and chronic inflammation in cytokine and hormone release
80 A.M. Mancha-Ramirez and T.J. Slaga
Excess dietary energy is stored in the adipocytes, and as this process occurs,
cytokines are released along with adipocyte-related hormones which affect systemic
vasculature [28, 106, 115]. Of the cytokines released via the process described
above, some of the most notable are angiogenin, vascular endothelial growth factor,
endostatin, tumor necrosis factor-a, and interleukin (IL)-6 [28, 106, 115].
This release of cytokines that takes place results in the activation of various
signaling pathways including nuclear transcription factor jB (NFjB) [8, 112].
NFjB is a ubiquitous transcription factor that normally resides in the cytoplasm, but
when activated, is translocated to the nucleus, where it induces transcription of
many genes [2]. In terms of obesity and NFjB, the obese state in general is
associated with increased systemic inflammatory cytokine production and chronic
activation of NFjB [55, 71]. In addition to its role in metabolic disorders, NFjB is
also upregulated in a multitude of human cancers and contributes to tumor pro-
motion [19, 47, 62, 72, 109, 110]. The activated form of NFjB is capable of
mediating cancer, atherosclerosis, diabetes, arthritis, and many other inflammatory
diseases [2]. Specifically, the NFjB pathway is one of the most important signaling
pathways involved in immunity, inflammation, proliferation, and anti-apoptotic
defenses. In terms of cancer, inhibition of NFjB leads to downregulation of pro-
teins involved in anti-apoptotic defense mechanisms utilized by cancer cells such as
Bcl-2, Bcl-xL, and c-FLIP; this process thereby promotes apoptotic cell death in
cancer cells [58]. The activation of IKKb plays a major role in inflammation
induced tumor promotion and progression [126]. Experimental studies have shown
the NFjB activity is necessary for the formation of a number of tumor types [38].
Increased NFjB activation may provide a link between obesity, diabetes, and a
range of tumor types [90, 91, 113].
It is well known that AMP-activated protein kinase (AMPK) is a fuel-sensing
enzyme that is activated by changes in the AMP/ATP ratio. The role of this enzyme
is to increase cellular ATP generation and diminish ATP use for less critical pro-
cesses depending upon the situation [93]. In addition to glucose transport, lipid and
protein synthesis, and fuel metabolism, AMPK is also responsible for regulating a
myriad of other physiological processes including but not limited to cellular growth
and proliferation, mitochondrial function, and factors linked to insulin resistance,
including inflammation, oxidative and ER stress, and autophagy [93]. AMPK
restores ATP levels by inhibiting anabolic processes like fatty acid synthesis and
gluconeogenesis and activating catabolic processes like fat oxidation and glucose
uptake [117]. Significant evidence in the literature supports a definite link between
dysregulation of AMPK and insulin resistance in both rodents and humans. In
addition, common antidiabetic drugs, as well as insulin-sensitizing hormones like
adiponectin, function via AMPK [23, 76, 121, 124, 136]. It has also been shown
that AMPK suppresses NFjB activities in different systems through a variety of
mechanisms, and antidiabetic drugs have also shown NFjB-suppressing properties
as a consequence of AMPK activation [9, 94, 133]. As many studies have shown
that calorie restriction can prevent tumor formation in various mouse models of
cancer, the link between obesity and cancer is one that is well represented in the
literature. Calorie restriction models have prevented tumor formation in brain,
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 81
colon, breast, prostate, and chemically mediated skin cancer [11, 12, 24, 69, 102]. It
has been suggested that the anticancer effects of calorie restriction and exercise may
be mediated by the energy sensor AMPK [67].
Additional mechanistic links between metabolic syndrome and cancer include as
we would expect, many of the cell signaling pathways related to inflammation such
as TNF-a. Additionally, interleukin-6 (IL-6) is also shown to be elevated in obesity
and is positively correlated with BMI [50]. Increased IL-6 levels, as seen in obesity
models, leads to induced JAK-STAT3 signal transduction and stimulates cell
proliferation, differentiation, and metastasis [21, 125]. Data related to insulin
resistance and mechanistic links to cancer suggest that insulin resistance may lead
to a poorer response to cancer treatment and possibly a more aggressive phenotype
in patients with preexisting diabetes [66].
Table 4.1 Various medicinal plants containing ursolic acid (adapted from [6])
Botanical name Common name
Ocimum sanctum L. Holy Basil (Tulsi)
Eriobotrya japonica Loquat
Vaccinium macrocarpon Cranberry
Harpagophytum procumbens Devil’s Claw
DC
Sambucus nigra L. Elder flowers (European variety)
Mentha piperita L. Peppermint leaves
Eugenia jambolana Java plum
Perilla frutescens Beefsteak plant
Apocynaceae Lavender
Lavandula augustifolia Mill.
Origanum vulgare L. Oregano
Malus domestica Different apples
Melissa officinalis Lemon balm
Rosmarinus officinalis Rosemary
Plantago major Greater plantain
Thymus vulgaris L. Thyme
Crataegus laevigata (Poir) Hawthorn
DC
Coffea arabica Coffee
Calluna vulgaris Heather
Eucalyptus spp. Eucalyptus
[126]. Although the focus here is on chronic illnesses, cancer and obesity, UA has
also been shown to be effective in treatment of chronic diseases related to car-
diovascular conditions, atherosclerosis, and osteoporosis [126].
UA is a phytonutrient with great versatility and is capable of multiple biological
functions; however one potential drawback to its potential use is that despite low
toxicity, it has low water solubility [127]. To overcome this potential drawback and
improve upon its activity and bioavailability, many studies have been carried out in
order to identify structural modifications to achieve this goal. Specifically, UA
derivatives with modifications at the 3-OH and/or 17-COOH positions demonstrate
significant antitumor effects both in vitro and in vivo suggesting that these modi-
fications can enhance the bioavailability of UA [13, 26, 80]. Additionally, structural
studies have also shown that modification of UA with a piperazine group, specif-
ically a piperazine moiety at the C-28 position, can elicit a better inhibitory effect on
cancer cell growth [22, 100]. Overall, of the many reported UA analogs which have
been developed to date, some of the most important modifications include those on
the positions of C-3/C-28, C-11, C-17, and C-28, and modifications on C-2/C-3
positions and ring A [16].
Although there have not been any detailed studies on pure UA and its absorption
and distribution, one study investigating the intestinal uptake of UA (from the
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 83
containing plants have anti-bacterial and antifungal activity; in fact, UA has been
shown to have antimicrobial activity against several strains of staphylococci [4,
130]. Finally, UA is a potent inhibitor of components of metabolic syndrome such
as hypertension, triglyceride levels, and accumulation of inflammatory monocytes
and associated atherosclerosis [41, 42, 108, 114].
ultimately lead to cellular dysfunction [30]. Animal studies showed that treatment
with UA was able to decrease liver damage caused by oxidative stress inducing
chemicals such as carbon tetrachloride [65]. Moreover, UA, and other triterpenoids
were also able to increase the activities of the antioxidant enzymes superoxide
dismutase and glutathione peroxidase [68, 118]. Another significant complication
of diabetes is diabetic nephropathy, and therefore UA was tested to determine what
effect it would have on renal function in streptozotocin-induced diabetes. The study
looking at diabetic nephropathy found that UA significantly prevented biochemical
and histopathologic changes in the kidneys associated with diabetes, and lowered
NF-jB activity and renal oxidative stress levels [63]. Finally, phase 2 clinical
studies evaluating the effect of UA on insulin sensitivity and metabolic syndrome
were carried out in 2015 but the results have not yet been published. Despite the
fact that recent studies have demonstrated that UA does exert an antiobesity effect,
the mechanisms of action are still under investigation. Currently, several of the
proposed mechanisms and targets include inhibition of PTP1B, lipolysis stimulation
via cAMP-dependent PKA pathway modulation, modulation of the LKB1/AMPK
pathway, and regulation of adipogenic differentiation [42, 57, 82, 89, 120].
With regards to cancer, UA has been studied in great detail. However, the precise
mechanisms of the many anticancer effects of UA and corresponding molecular
targets for these capabilities are still under investigation. UA has been shown to
have a variety of anticancer capabilities, including the ability to suppress prolif-
eration of several different types of tumor cells, induce apoptosis, and inhibit tumor
promotion, metastasis, and angiogenesis in cancer animal models [98]. Several
mechanisms which have been elucidated for the above mentioned anticancer
abilities of UA thus far and include the ability to suppress multiple cell signaling
pathways such as growth factor receptor activation (EGFR), signaling through
IKK/NF-jB, Akt/mTOR, Cox-2, STAT3, MMP9, and VEGF [97, 126, 131].
papilloma and carcinoma cell lines have indicated that the cytotoxic effects of UA
can be enhanced with p-glycoprotein inhibitors [44].
Cranberry-derived UA showed potent anti-proliferative activities against HepG2
liver cancer cells and MCF7 breast cancer cells [35]. UA-mediated apoptosis of
human bladder cancer cells was strongly suppressed by AMPK knockdown [135].
Additionally, UA reduced the viability of human colon cancer cell lines [27].
UA has also shown great promise in breast cancer studies as well, with an ability
to strongly decrease viability of a breast cancer cell line with an associated inhi-
bition of NFjB, as well as, a decrease size of breast cancer xenografts in murine
models [129].
Similar in vitro effects of UA have been seen in prostate cancer models as well
[99]. Notably, it was found that UA was able to suppress NFjB activation induced
by numerous carcinogens, such as TNFa, phorbol ester, okadaic acid, H2O2, and
cigarette smoke [103]. Inhibition of NFjB activation was mediated via IKK kinase
activation, IjBa phosphorylation and degradation, p65 phosphorylation, p65
nuclear translocation, and NFjB dependent reporter gene expression [98]. Results
from multiple myeloma in vitro studies showed that UA was able to inhibit both
constitutive and IL-6-inducible STAT3 activation, which was mediated through the
inhibition of upstream kinases C-SRC, JAK1, JAK2, and ERK1/2 [85]. Moreover,
they also showed that UA could induce expression of the SHP-1 protein, while
knockdown of SHP-1 via RNA interference was able to suppress the induction of
SHP-1 and reverse the inhibition of STAT3 activation. The results from the Pathak
group suggest a critical role of SHP-1 in the mechanism of action of UA, and show
that it can be effective in suppressing important inflammatory networks including
NFjB, STAT3, and AKT [85].
In addition to the extensive work that has been carried out investigating the efficacy
of UA in in vitro cancer models, significant evidence supporting the versatility and
usefulness of UA has also been amassed in in vivo studies as well. For example, the
effect of UA was evaluated on early stages of skin tumorigenesis utilizing a
two-stage carcinogenesis SENCAR mouse model, and found topical application of
UA alone and/or in combination with calcium D-glutarate during the promotion
stage was able to inhibit tumor multiplicity and tumor incidence [52]. Results from
this study showed that UA is a potent inhibitor of skin tumor promotion and
inflammatory signaling in skin cancer and potentially other epithelial cancers in
humans as well [52]. UA isolated from P. frutescens was also evaluated in a
two-stage skin carcinogenesis mouse model, and was found to significantly inhibit
skin tumor promotion by the tumor promoting agent, TPA [17, 18]. Specifically,
pretreatment with UA resulted in a 42 % inhibition of papilloma formation [17, 18].
Furthermore, when UA was used in combination with resveratrol, a phytochemical
present in grapes, berries, peanuts, and red wine, the combination was found to
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 87
have a greater inhibitory effect on skin tumor promotion by TPA in a mouse model
than with either agent used independently [17, 18]. Mechanistic studies into the
enhanced inhibitory effects observed in the UA + resveratrol combination showed
an upregulation of tumor suppressor genes such as p21 and PDCD4 as well as a
dramatic increase of p-AMPKa and its downstream target p-Ulk1-ser555 [17, 18].
In a prostate cancer DU145 xenograft nude mouse model, one group analyzed
whether UA could inhibit the growth of prostate cancer and found that UA was able
to significantly suppress the tumor growth in vivo following 6 weeks of treatment
[99]. Also noteworthy in this xenograft prostate cancer study was that at the end-
point of the experiment, there was no significant change in body weight of
UA-treated mice suggesting that UA is a nontoxic compound when administered to
mice. Results from this in vivo prostate cancer mouse model showed a substantial
decrease in VEGF expression and an increase in caspase-3 expression in
UA-treated mice, suggesting that perhaps UA is functioning as an anti-angiogenic
and pro-apoptotic agent against prostate cancer in this model [99]. Additionally,
UA, when administered by gavage, also decreased incidence of chemically induced
preneoplastic lesions in rat colon [27] (Fig. 4.3).
Finally, a phase I trial was carried out in 2015 to evaluate the multiple-dose
safety and antitumor activity of UA liposomes (UAL) in patients with advanced
solid tumors. UA liposome is a new antitumor drug that has significant potential
therapeutic value as it utilizes liposomes to overcome the poor solubility of UA,
increase the therapeutic efficiency, reduce the side effects, and enhance the
bioavailability of the drug. Results from this clinical trial demonstrate that UAL
treatment of patients with advanced solid tumors via multiple-dose and consecutive
Fig. 4.3 An overview of the many oncogenic targets UA has been shown to modulate
88 A.M. Mancha-Ramirez and T.J. Slaga
14-day intravenous infusion every 21 days was safe. Additionally, the prelimi-
nary antitumor activity of UAL was evaluated for the first time in a clinical setting
and their results indicate UAL may be able to potentially improve patient
remission [87].
4.6 Conclusion
As obesity, metabolic syndrome, and cancer have been shown to have similar
mechanistic links, this presents a useful avenue for novel preventive and therapeutic
strategies aimed at fighting these chronic diseases. Dysregulation of important cell
signaling pathways occurs during the constant state of inflammation during obesity,
and this has been linked to an increased overall risk of certain cancers such as breast
and colorectal cancer. Moreover, as discussed earlier, patients with preexisting
conditions brought on by obesity, such as insulin resistance, tend to respond more
poorly to traditional chemotherapeutic agents than otherwise healthy individuals.
As cancer is a multifactorial disease, it is vital to investigate novel therapeutic
compounds that exhibit efficacy against key pathways often dysregulated in cancer
pathogenesis. Due to the unique nature of each individual cancer development,
multi-targeting therapies from natural resources are vital for targeting cancer
development and progression. Triterpenoids such as UA offer unique and novel
drug platforms for development due to the fact that they have been shown to target
critical inflammatory proteins for the prevention and treatment of cancer. As these
phytonutrient have been shown to be nontoxic, it is their safety and ability to affect
various key targets that makes compounds like UA very desirable as a starting point
for future drug development. It is quite possible that triterpenes such as UA could
be used in conjunction with commonly used chemotherapeutic agents to allow for
perhaps a decrease in the chemotherapeutic dose, less adverse effects of the
chemotherapeutic agent, or even synergistic effects between UA and the drug.
While there is significant data in the literature to support an important role for UA
in cancer prevention/therapy, future clinical investigations are needed to further
validate these findings. Here, we have discussed in detail the various ways in which
UA has shown great potential as an anticancer agent in many different types of
cancer; many of these results have significant clinical implications as UA has been
shown to target key cancer pathways.
UA is not only capable of acting as an anticancer compound, but it also has been
shown to have versatile antiobesity properties which we have described in detail.
These effects appear to be partially mediated by inhibition of NFjB and/or acti-
vation of AMPK, but due to the many various effects of UA seen in both cancer and
obesity, the precise mechanisms of action for these effects are still unclear.
Although diet and exercise plus a highly controlled blood glucose level is currently
the recommended treatment for obesity and type II diabetes, one of the major
drawbacks of this standard of care is of course compliance. Where patients may
exhibit poor compliance with a recommended course of diet and exercise,
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 89
References
1. Aggarwal B, Van Kuiken ME, Iyer LH, Harijumar KB, Sung B (2009) Molecular targets of
nutraceuticals derived from dietary spices: potential role in suppression of inflammation and
tumorigenesis. Exp Biol Med 234:825–849
2. Aggarwal B, Shishodia S (2004) Suppression of the nuclear factor-jB activation pathway by
spice-derived phytochemicals. Ann NY Acad Sci 1030:434–441
3. Aggarwal B, Shishodia S (2006) Molecular targets of dietary agents for prevention and
therapy of cancer. Biochem Pharmacol 71:1397–1421
4. Anisimov MM, Shcheglov VV, Strigina LI et al (1979) Chemical structures and antifungal
activity of a number of triterpenoids. Biol Bull Acad Sci USSR 6:464–468
5. Aridiacono B, Iiritano S, Nocera A, Possidente K, Nevolo M, Ventura V, Foti D, Chiefari E,
Brunetti A (2012) Insulin resistance and cancer risk: an overview of the pathogenic
mechanisms. Exp Diabetes Res 2012:1–12
6. Babalola I, Shode F (2013) Ubiquitous ursolic acid: a potential pentacyclic triterpene natural
product. J Pharmacogn Phytochem 2:214–222
7. Balunas MJ, Kinghorn AD (2005) Drug discovery from medicinal plants. Life Sci 78:
431–441
8. Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H (2006) Recent advances in
the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw
1:4–12
9. Bess E, Fisslthaler B, Fromel T, Fleming I (2011) Nitric oxide-induced activation of the
AMP-activated protein kinase a2 subunit attenuates IjB kinase activity and inflammatory
responses in endothelial cells. PLoS One 6:e20848
10. Bessaoud F, Daures J (2008) Dietary factors and breast cancer risk: a case control study
among a population in southern France. Nutr Cancer 60:177–187
90 A.M. Mancha-Ramirez and T.J. Slaga
11. Birt DF, Pelling JC, White LT, Dimitroff K, Barnett T (1991) Influence of diet and calorie
restriction on the initiation and promotion of skin carcinogenesis in SENCAR mouse model.
Cancer Res 51:1851–1854
12. Blando J, Moore T, Hursting S, Jiang G, Saha A, Beltran L (2011) Dietary energy balance
modulates prostate cancer progression in Hi-Myc mice. Cancer Prev Res 4:2002–2014
13. Brenner DA (2011) Molecular pathogenesis of liver fibrosis. Trans Am Clin Climatol Assoc
120:361–368
14. Camer D, Yu Y, Szabo A, Huang XF (2014) The molecular mechanisms underpinning the
therapeutic properties of oleanolic acid, its isomer and derivatives for type 2 diabetes and
associated complications. Mol Nutr Food Res 58:1750–1759
15. Campbell PT, Jacobs ET, Ulrich CM, Figueiredo JC, Poynter JN, McLaughlin JR et al
(2010) Case–control study of overweight, obesity, and colorectal cancer risk, overall and by
tumor microsatellite instability status. J Natl Cancer Inst 6:391–400
16. Chen H, Gai Y, Wang A, Zhou X, Zheng Y, Zhou J (2015) Evolution in medicinal chemistry
of ursolic acid derivatives as anticancer agents. Eur J Med Chem 92:648–655
17. Cho J, Rho O, Junco J, Carbajal S, Siegel D, Slaga T, DiGiovanni J (2015) Effect of
combined treatment with ursolic acid and resveratrol on skin tumor promotion by 12-O-
tetradecanoylphorbol-13-acetate. Cancer Prev Res 8:817–825
18. Cho J, Tremmel L, Rho O, Camelio A, Siegel D, Slaga T, DiGiovanni J (2015) Evaluation of
pentacyclic triterpenes found in Perilla frutescens for inhibition of skin tumor promotion by
12-O-tetradecanoylphorbol-13-acetate. Oncotarget 6:39292–39306
19. Cogswell PC, Guttridge DC, Funkhouse WK, Baldwin AS (2000) Selective activation of
NF-jB subunits in human breast cancer: potential roles for NF-jB 2/p52 and for Bcl-3.
Oncogene 19:1123–1131
20. Cong LN, Chen H, Li YH, Zhou LX et al (1997) Physiological role of Akt in
insulin-stimulated translocation of GLUT4 in transfected rat adipose cells. Mol Endocrinol
11:1881–1890
21. Darnell JE Jr, Kerr IM, Stark GR (1994) Jak-STAT pathways and transcriptional activation
in response to IFNs and other extracellular signaling proteins. Science 264:1415–1421
22. Dong HY, Yang X, Xie JJ, Xiang LP, Li YF, Ou MR, Chi T, Liu ZH, Yu SH, Gao Y,
Chen JZ, Shao JW, Jia L (2015) UP12, a novel ursolic acid derivative with potential for
targeting multiple signaling pathways in hepatocellular carcinoma. Biochem Pharmacol
93:151–162
23. Fediuc S, Pimenta AS, Gaidhu MP, Ceddia RB (2008) Activation of AMP-activated protein
kinase, inhibition of pyruvate dehydrogenase activity, and redistribution of substrate
partitioning mediate the acute insulin-sensitizing effects of troglitazone in skeletal muscle
cells. J Cell Physiol 215:392–400
24. Fernandes G, Chandrasekar B, Troyer DA, Venkatraman JT, Good RA (1995) Dietary lipids
and calorie restriction affect mammary tumor incidence and gene expression in mouse
mammary tumor virus/v-Ha-ras transgenic mice. Proc Natl Acad Sci 92:6494–6498
25. Freedland SJ, Platz EA (2007) Obesity and prostate cancer: making sense out of apparently
conflicting data. Epidemiol Rev 29:88–97
26. Friedman SL (2008) Mechanisms of hepatic fibrogenesis. Gastroenterology 134(6):
1655–1669
27. Furtado RA, Rodrigues EP, Araujo FR, Oliveira WL, Furtado MA et al (2008) Ursolic acid
and oleanolic acid suppress preneoplastic lesions induced by 1,2-dimethylhydrazine in rat
colon. Toxicol Pathol 36:576–580
28. Galic S, Oakhill JS, Steinberg GR (2009) Adipose tissue as an endocrine organ. Mol Cell
Endocrinol 285:115–122
29. Gan KH, Lin CN (1988) Studies on the constituents of Formosan gentianaceous plants. XI.
Constituents of Gentiana flavor-maculata and tripterospermum Taiwanese and the
cento-hepatotoxic activities of ursolic acid derivatives. Chin Pharmaceut J 40:77–84
30. Gao D, Li Q, Li Y, Liu Z et al (2009) Antidiabetic and antioxidant effects of oleanolic acid
from Ligustrum lucidum Ait in alloxan-induced diabetic rats. Phytother Res 23:1257–1262
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 91
31. Garrido JC, Cevallos GAC, Siciliano LG et al (2012) Acute and subacute toxicity (28 day) of
a mixture of ursolic acid and oleanolic acid obtained from Bouvard interminfolia in mice.
Bull Latinoam Caribe Plantas Medic Aromat 11(1):91–102
32. Guarente L (2006) Sirtuins as potential targets for metabolic syndrome. Nature 444:868–874
33. Hajduch E, Alessi DR, Hemmings BA, Hundal HS (1998) Constitutive activation of protein
kinase B alpha by membrane targeting promotes glucose and system A amino acid transport,
protein synthesis, and inactivation of glycogen synthase kinase 3 in L6 muscle cells. Diabetes
47:1006–1013
34. He Y, Li Y, Zhao T, Wang Y, Sun C (2013) Ursolic acid inhibits adipogenesis in 3T3-L1
adipocytes through LKB1/AMPK pathway. PLoS One 8:1–12
35. He X, Liu RH (2006) Cranberry phytochemicals: isolation, structure elucidation, and their
antiproliferative and antioxidant activities. J Agric Food Chem 54:7069–7074
36. He W, Shi F, Zhou Z-W, Li B, Zhang K, Zhang X et al (2015) A bioinformatic and
mechanistic study elicits the antifibrotic effect of ursolic acid through the attenuation of
oxidative stress with the involvement of ERK, PI3K/Akt, and p38 MAPK signaling pathways
in human hepatic stellate cells and rat liver. Drug Des Dev Ther 9:3989–4104
37. Hill JO, Wyatt HR, Reed GW, Peters JC (2003) Obesity and the environment: where do we
go from here? Science 299:853–855
38. Huang S, Pettaway CA, Uehara H, Bucana CD, Fidler IJ (2001) Blockade of NF-jB activity
in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and
metastasis. Oncogene 31:4188–4197
39. Hunan Med Inst (1975) Pharmacological studies of hepatoprotective compounds from
Swertia mileenses. Trad Med (Zhang Chao Yao) 6:47–62
40. Jager S, Trojan H, Kopp T, Laszczyk MN, Scheffler A (2009) Pentacyclic triterpene
distribution in various plants—rich sources for a new group of multi-potent plant extracts.
Molecules 14:2016–2031
41. Jang SM, Yee ST, Choi J et al (2009) Ursolic acid enhances the cellular immune system and
pancreatic b-cell function in streptozotocin-induced diabetic mice fed a high-fat diet. Int
Immunopharmacol 9(1):113–119
42. Jayaprakasam B, Olson LK, Schutzki RE, Tai MH, Nair M (2006) Amelioration of obesity
and glucose intolerance in high-fat-fed C57BL/6 mice by anthocyanins and ursolic acid in
cornelian cherry (Cornus mas). J Agric Food Chem 54:243–248
43. Jedrychowski W, Maugeri U, Popiela T, Kulig J, Sochacka-Tatara E, Pac A, Sowa A,
Musial A (2010) Case–control study on beneficial effect of regular consumption of apples on
colorectal cancer risk in a population with relatively low intake of fruits and vegetables. Eur J
Cancer Prev 19:42–47
44. Junco J, Mancha A, Malik G, Wei S, Kim DJ, Liang H, Slaga TJ (2013) Resveratrol and p-
glycoprotein inhibitors enhance the anti-skin cancer effects of ursolic acid. Mol Cancer Res
11:1521–1529
45. Junco J, Mancha-Ramirez A, Malik G, Wei S, Kim DJ, Liang H, Slaga TJ (2015) Ursolic
acid and resveratrol synergize with chloroquine to reduce melanoma cell viability. Melanoma
Res 25:103–112
46. Jung SH, Ha YJ, Shim EK, Choi SY et al (2007) Insulin-mimetic and insulin-sensitizing
activities of a pentacyclic triterpenoid insulin receptor activator. Biochem J 403:243–250
47. Karin M (2009) NF-jB as a critical link between inflammation and cancer. Cold Spring Harb
Perspect Biol 1:a000141
48. Kelley DE, Wing R, Buonocore C, Sturis J, Polonsky K, Fitzsimmons M (1993) Relative
effects of calorie restriction and weight loss in noninsulin-dependent diabetes mellitus. J Clin
Endocrinol Metab 5:1287–1293
49. Kelloff GJ, Crowell JA, Steele VE, Lubet RA, Malone WA, Boone CW, Kopelovich L, Et
Hawk, Lieberman R, Lawrence JA, Ali I, Viner JL, Sigman CC (2000) Progress in cancer
chemoprevention: development of diet-derived chemopreventive agents. J Nutr 130:
467S–471S
92 A.M. Mancha-Ramirez and T.J. Slaga
50. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G (2001) Adipose tissue tumor
necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J
Physiol 280:E745–E751
51. Kloppel E, Sinzato YK, Damasceno DC, Volpato GT, Campos KE (2015) Effect of maternal
obesity on insulin action in male adult offspring rats. Diabetol Metab Syndr 7:A127
52. Kowalczyk MC, Junco J, Kowalczyk P, Tostykh O, Hanausek M, Slaga TJ, Walaszek Z
(2013) Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice. Int J
Oncol 43:911–918
53. Kowalczyk MC, Walaszek Z, Kowalczyk P et al (2009) Differential effects of several
phytochemicals and their derivatives on murine keratinocytes in vitro and in vivo:
implications for skin cancer prevention. Carcinogenesis 30(6):1008–1015
54. Kunkel SD, Elmore CJ, Bongers KS, Ebert SM, Fox DK, Dyle MC, Bullard SA, Adams CM
(2012) Ursolic acid increases skeletal muscle and brown fat and decreases diet-induced
obesity, glucose intolerance and fatty liver disease. PLoS One 7:1–8
55. Lappas M, Yee K, Permezel M, Rice GE (2005) Sulfasalazine and BAY 11-7082 interfere
with the nuclear factor-jB and kinase pathway to regulate the release of proinflammatory
cytokines from human adipose tissue and skeletal muscle in vitro. Endocrinology 3:
1491–1497
56. Lee J, Yee ST, Kim JJ, Choi MS, Kwon EY, Seo KI, Lee MK (2010) Ursolic acid
ameliorates thymic atrophy and hyperglycemia in streptozotocin-nicotinamide-induced
diabetic mice. Chem Biol Interact 188:635–642
57. Li Y, Xing D, Chen Q, Chen W (2010) Enhancement of chemotherapeutic agent-induced
apoptosis by inhibition of NF-jB using ursolic acid. Int J Cancer 127:462–473
58. Li-Weber M (2010) Targeting apoptosis pathways in cancer by Chinese medicine. Cancer
Lett 332:304–312
59. Liao Q, Yang W, Jia Y et al (2005) LC-MS Determination and pharmacokinetic studies of
ursolic acid in rat plasma after administration of the traditional Chinese medicinal preparation
Lu–Ying extract. Yakugaku Zasshi 125(6):47–62
60. Lin CN, Chung MI, Gan KH (1988) Novel anti-hepatotoric principles of Solanun incanum L.
Planta Med 54:222
61. Lin ZH, Zhang Y, Zhang YN, Shen H et al (2008) Oleanolic acid derivative NPLC441
potently stimulates glucose transport in 3T3-L1 adipocytes via a multi-target mechanism.
Biochem Pharmacol 76:1251–1262
62. Lind DS, Hochwald SN, Malaty J, Rekkas S, Hebig P, Mishra G (2001) Nuclear factor-jB is
upregulated in colorectal cancer. Surgery 130:363–369
63. Ling C, Jinping L, Xia L, Renyong Y (2013) Ursolic acid provides kidney protection in
diabetic rats. Curr Ther Res 75:59–63
64. Liu J (1995) Pharmacology of oleanolic acid and ursolic acid. J Ethnopharmacol 49:57–68
65. Liu J, Liu Y, Mao Q, Klaassen CD (1994) The effects of 10 triterpenoid compounds on
experimental liver injury in mice. Fundam Appl Toxicol 22:34–40
66. Louie SM, Roberts LS, Nomura DK (2013) Mechanisms linking obesity and cancer.
Biochim Biophys Acta 1831:1499–1508
67. Lupertz R, Chovolou Y, Kampkotter A, Watjen W, Kahl R (2008) Catalase overexpression
impairs TNF-a induced NF-jB activation and sensitizes MCF-7 cells against TNF-a. J Cell
Biochem 103:1497–1511
68. Ma XH, Zhao YC, Yin L et al (1986) Studies on the preventive and therapeutic effects of
ursolic acid on acute hepatic injury in rats. Acta Pharm Sin 21:332–335
69. Mai V, Colbert LH, Berrigan D, Perkins SN, Pfeiffer R, Lavigne JA (2003) Calorie
restriction and diet composition modulate spontaneous intestinal tumorigenesis in Apc(Min)
mice through different mechanisms. Cancer Res 63:1752–1755
70. Martin C, Zhang Y, Tonelli C, Petroni K (2013) Plants, diet, and health. Annu Rev Plant Biol
64:19–46
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 93
91. Rial NS, Choi K, Nguyen T, Snyder B, Slepian MJ (2012) Nuclear factor jB: a novel cause
for diabetes, coronary artery disease and cancer initiation and promotion? Med Hypotheses
78:29–32
92. Rodriguez C, Freedland SJ, Deka A, Jacobs EJ, McCullough ML, Patel AV et al (2007)
Body mass index, weight change, and risk of prostate cancer in the Cancer Prevention
Study II Nutrition Cohort. Cancer Epidemiol Biomarkers Prev 16:63–69
93. Ruderman NB, Carling D, Prentki M, Cacicedo JM (2013) AMPK, insulin resistance, and the
metabolic syndrome. J Clin Investig 123:2764–2772
94. Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J (2008) Terpenoids:
natural inhibitor sof NF-jB signaling with anti-inflammatory and anticancer potential. Cell
Mol Life Sci 65:2979–2999
95. Sarkar FH, Li YW (2007) Targeting multiple signal pathways by chemopreventive agents for
cancer prevention and therapy. Acta Pharmacol Sin 28:1305–1315
96. Senn HJ, Kerr D (2011) Chronic non-communicable diseases, the European Chronic Disease
Alliance—and cancer. Ann Oncol 22:248–249
97. Shanmugam M, Dai X, Kumar AP, Tan B, Sethi G, Bishayee A (2013) Ursolic acid in
cancer-prevention and treatment: molecular targets, pharmacokinetics and clinical studies.
Biochem Pharmacol 85:1579–1587
98. Shanmugam M, Nguyen A, Kumar AP, Tan B, Sethi G (2012) Targeted inhibition of tumor
proliferation, survival, and metastasis by pentacyclic triterpenoids: potential role in
prevention and therapy of cancer. Cancer Lett 320:158–170
99. Shanmugam M, Rajendran P, Li F, Nema T, Vali S, Abbasi T, Kapoor S, Sharma A,
Kumar A, Ho P, Hui K, Sethi G (2011) Ursolic acid inhibits multiple cell survival pathways
leading to suppression of growth of prostate cancer xenograft in nude mice. J Mol Med
89:713–727
100. Shao JW, Dai YC, Xue JP, Wang JC, Lin FP, Guo YH (2011) In vitro and in vivo anticancer
activity evaluation of ursolic acid derivatives. Eur J Med Chem 46:2652–2661
101. Shaw JE, Sicree RA, Zimmet PZ (2010) Global estimates of the prevalence of diabetes for
2010 and 2030. Diabetes Res Clin Pract 87:4–14
102. Shelton LM, Huysentruyt LC, Mukherjee P, Seyfreid TN (2010) Calorie restriction as an
anti-invasive therapy for malignant brain cancer in the MV mouse. ASN Neurosci 2:e00038
103. Shishodia S, Majumdar S, Banerjee S, Aggarwal B (2003) Ursolic acid inhibits nuclear
factor-jB activation induced by carcinogenic agents through suppression of IjBa kinase and
p65 phosphorylation: correlation with down-regulation of cyclooxygenase 2, matrix
metalloproteinase 9, and cyclin D1. Cancer Res 63:4375–4383
104. Shuckla B, Viser S, Patnaik GK et al (1992) Hepatoprotective activity in the rat of ursolic
acid isolated from Eucalyptus hybrid. Phytother Res 6:74–79
105. Siddiqui AA (2011) Metabolic syndrome and its association with colorectal cancer: a review.
Am J Med Sci 3:227–231
106. Silha JV, Krsek M, Suchurda P, Murphy LJ (2005) Angiogenic factors are elevated in
overweight and obese individuals. Int J Obes 29:1308–1314
107. Singh GB, Singh S, Bani S et al (1992) Anti-inflammatory activity of oleanolic acid in rats
and mice. J Pharm Pharmacol 44:456–458
108. Somova LO, Nadar A, Rammanan P et al (2003) Cardiovascular antihyperlipidemic and
antioxidant effects of oleanolic and ursolic acids in experimental hypertension.
Phytomedicine 10(2–3):115–121
109. Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM et al (1997) Aberrant
nuclear factor-jB/Rel expression and the pathogenesis of breast cancer. J Clin Invest
100:2952–2960
110. Suh J, Rabson AB (2004) NF-jB activation in human prostate cancer: important mediator or
epiphenomenon? J Cell Biochem 91:100–117
111. Sun T, Wang Q, Yu ZG, Zhang Y et al (2007) Hyrtiosal, a PTP1B inhibitor from the marine
sponge Hyrtios erectus, shows extensive cellular effects on PI3K/AKT activation, glucose
transport, and TGF b/Smad2 signaling. Chem Biol Chem 8:187–193
4 Ursolic Acid and Chronic Disease: An Overview of UA’s Effects … 95
134. Zhang WL, Zhu L, Jiang JG (2014) Active ingredients from natural botanicals in the
treatment of obesity. Obes Rev 15:957–967
135. Zheng QY, Jin FS, Yao C, Zhang T, Zhang GH, Ai X (2012) Ursolic acid-induced
AMP-activated protein kinase (AMPK) activation contributes to growth inhibition and
apoptosis in human bladder cancer T24 cells. Biochem Biophys Res Commun 419:741–747
136. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J et al (2001) Role of
AMP-activated protein kinase in mechanism of metformin action. J Clin Investig 108:
1167–1174
Chapter 5
Tocotrienol and Its Role in Chronic
Diseases
Keywords Bone Cancer Cholesterol Diabetes Glucose Osteoporosis
Tocopherol Tocotrienol Vitamin E
5.1 Tocotrienol
Oxidants such as reactive oxygen species (ROS) and reactive nitrogen species
(NOS) are essential for defence against infectious agents, for cell signalling and for
redox homeostasis [170]. They are balanced by endogenous and exogenous
antioxidants in our body, such as antioxidant enzymes, vitamins, thiols and glu-
tathione. Oxidative stress occurs when the production of oxidants overwhelms the
production of antioxidants in our body [152, 153]. Free radical species are capable
of damaging macromolecules such as carbohydrate, protein and DNA in the body
and generate more free radicals, thereby forming a vicious cycle [48]. Many adverse
health conditions, such as neurodegeneration [12], diabetes mellitus [9],
100 K.-Y. Chin et al.
cardiovascular disease [82] and osteoporosis [20], have been linked to oxidative
stress. This highlights a possible role of tocotrienol, a strong antioxidant, in pre-
venting these oxidative stress-related diseases.
Both tocotrienol and tocopherol exert their free radical scavenging activity by
donating phenolic hydrogen at the sixth position of the chromanol ring [113].
Tocotrienol has been shown to be a superior membrane antioxidant compared to its
tocopherol counterpart [161]. This is due to the uniform distribution of tocotrienol
in the membrane and its stronger ability to disorder membrane lipids, thus
enhancing its recycling efficiency [145]. In studies using lipid peroxidation in
microsomes extracted from rat liver, Serbinova et al. showed that the antioxidant
effect of alpha-tocotrienol was 40–60 times stronger than alpha-tocopherol [145].
They attributed the superior antioxidant effect of tocotrienol to its higher recycling
efficacy from chromanoxyl radicals, uniform distribution in the membrane bilayers
and stronger disordering of membrane lipids structure, resulting in a higher efficacy
in its interaction with free radicals [145]. Palozza et al. showed that tocotrienol
isomers inhibited lipid peroxidation, ROS production and heat shock protein
expression in rat liver microsomal membrane and in RAT-1 immortalized fibrob-
lasts challenged with free radicals [121]. The effectiveness of delta-tocotrienol was
found to be greater than gamma-tocotrienol, and similar between gamma- and
alpha-tocotrienol [121]. It was proposed that decreased methylation of the chro-
manol ring, as in delta-tocotrienol, allowed the molecule to be better incorporated
into cell membranes [121]. Kamat et al. showed that palm tocotrienol significantly
reduced the lipid and protein peroxidation products in the microsomal extracts of
the brain and liver from rats [66, 67]. Among the individual isomers, they identified
that gamma-tocotrienol had the highest antioxidant efficacy compared to delta- and
alpha-tocotrienol [66, 67].
The antioxidant properties of tocotrienol were also demonstrated in cell culture
studies. Tan et al. showed that alpha-tocotrienol prevented the decrease of glu-
tathione and mitochondrial depolarization induced by hydrogen peroxide in hepa-
tocytes [166]. The increase of cellular ROS and malondialdehyde, a lipid
peroxidation product, induced by hydrogen peroxide and paracetamol in hepato-
cytes were also suppressed by alpha-tocotrienol. Nizar et al. showed that
gamma-tocotrienol prevented the apoptosis of primary osteoblast induced by
hydrogen peroxide [115]. In a subsequent experiment, they showed that this was
achieved by the preservation of cellular antioxidant defence system, such as
superoxide dismutase, catalase and glutathione peroxidase, and the reduction of
lipid peroxidation product in the treated cells [1].
Tocotrienol also exhibited its antioxidant effect in animal models. Nesaretnam
et al. demonstrated that long-term feeding of rats with diet containing palm oil rich
in tocotrienol significantly reduced the peroxidation potential of hepatic mito-
chondria and microsomes compared to corn oil control [106]. The initial rate of
lipid peroxidation was also found to be slower in the palm oil supplemented group
compared to the corn oil control [106]. Lee et al. showed that rats supplemented
with tocotrienol-rich fraction at 25 or 50 mg/kg body weight for 28 days and forced
to undergo a swimming endurance test showed higher liver superoxide dismutase,
5 Tocotrienol and Its Role in Chronic Diseases 101
catalase and glutathione peroxidase activity but lower liver lipid peroxidation and
reduced liver and muscle protein carbonyl levels compared to the vehicle and
alpha-tocopherol (25 mg/kg body weight) groups [79]. Supplementation of
alpha-tocotrienol (0.04 % weight diet) for one month in rats fed with vitamin
E-deficient diet reduced the level of hydroxyoctadecadienoic acid and isoprostane,
both markers of lipid peroxidation, in plasma and various organs of the rats [186].
The limited examples presented above showcased the antioxidant potential of
tocotrienol ex vivo and in vivo. The role of tocotrienol in suppressing oxidative
stress in relation to specific diseases will be discussed separately in the later
sections.
The mevalonate pathway is responsible for the synthesis of cholesterol and other
isoprenoids. The determining step in this multi-cascade pathway is the synthesis of
3-hydroxy-3-methyglutaryl-CoA (HMG-CoA) from acetyl-CoA via the enzyme
HMG-CoA reductase (HMGR). The expression of HMGR is in turn regulated by
sterol regulatory element-binding proteins (SREBPs), whereby the absence of sterol
isoprenoids in the cells upregulates its expression (reviewed by Buhaescu and
Izzedine [17]). Cholesterol is carried in lipoproteins with specific apolipoproteins
which direct the load to specific tissues. Excretion of cholesterol occurs in the form
5 Tocotrienol and Its Role in Chronic Diseases 103
Watkins et al., rats were fed with an atherogenic diet for 6 weeks and the treatment
groups received 50 mg gamma-tocotrienol and 500 mg alpha-tocopherol per kg
diet [179]. The treatment group showed lower plasma cholesterol, LDL, very–
low-density lipoprotein (VLDL), TG, malondialdehyde and fatty acid hydroper-
oxides [179]. Minhajuddin et al. demonstrated that rats on a 3-week atherogenic
diet showed significant reduction in TG, total and LDL cholesterol, malondialde-
hyde and conjugated dienes after supplementation with tocotrienol-rich fraction (8–
48 mg/kg) for 1 week [91]. The activity of HMGR was suppressed with athero-
genic diet, probably due to physiological feedback mechanism [91].
Supplementation of tocotrienol-rich fraction led to further reduction in the activity
of HMGR [91].
Using a chemically induced hypercholesterolemic model, Iqbal et al. showed
that supplementation of rice bran tocotrienol-rich fraction (10 mg/kg/day) signifi-
cantly suppressed the increase in plasma total and LDL cholesterol in rats admin-
istered with 7,12-dimethylbenz[alpha]anthracene [62]. It also suppressed the
activity and protein mass of hepatic HMGR [62]. Salman Khan et al. demonstrated
that Tocomin, a palm tocotrienol mixture, reduced the level of plasma lipoproteins,
cholesterol, apolipoprotein B, small dense LDL and LDL in inflammation-induced
hypercholesterolemia in Syrian hamsters [142]. In the same experiment using
computational modelling, they showed that the inhibition of HMGR was greater
with delta-tocotrienol, followed by gamma-, beta- and alpha-tocotrienol [142].
Raederstorff et al. compared the hypocholesterolemic effects of pure
gamma-tocotrienol and tocotrienol mixture in hamsters [134]. They found that both
gamma-tocotrienol (at 58 and 263 mg/kg) and tocotrienol mixture (at 263 mg/kg)
reduced plasma total and LDL cholesterol but TG and high density lipoprotein
(HDL) cholesterol were not affected [134]. Thus, the hypocholesterolemic effect of
pure gamma-tocotrienol was greater than the tocotrienol mixture [134]. Khor and
Ng showed that presence of alpha-tocopherol might attenuate the hypocholes-
terolemic effect of tocotrienol [74]. In male hamsters fed with diet containing
low-dose alpha-tocopherol (30 ppm), the activity of HMGR was inhibited [74].
However, diet containing high-dose alpha-tocopherol (81 ppm) significantly stim-
ulated the activity of HMGR [74]. On the other hand, in guinea pigs treated with
10 mg tocotrienol (i.p.), showed significant inhibition of HMGR activity but
mixture of 5 mg alpha-tocopherol and 10 mg tocotrienol caused less inhibition
[74]. This observation in turn validated the experiment of Qureshi et al. using
chickens [124]. In a further experiment using guinea pigs injected with palm olein
triglycerides containing either tocotrienol or alpha-tocopherol, Khor et al. indicated
that tocotrienol (1–5 mg) significantly reduced the activity of liver microsomal
HMGR and cholesterol-7-alpha-hydroxylase [73]. The inhibition by
alpha-tocopherol was less effective [73]. Furthermore, the inhibitory effect of
alpha-tocopherol was lost at 5 mg [73]. At a higher dose (50 mg), alpha-tocopherol
significantly elevated the activity of HMGR and reduced the inhibitory effects of
tocotrienol [73].
Effects of tocotrienol supplementation on lipid profile in humans were hetero-
geneous. This could be due to the differences in the composition of the tocotrienol
106 K.-Y. Chin et al.
mixture used, population studied, diet of the subjects, etc. In an earlier study using
palm vitamin E (containing 18 mg tocopherols and 42 mg tocotrienols), Tan et al.
showed that supplementation for 30 days lowered serum total and LDL cholesterol
but the TG and HDL cholesterol were not affected [167]. However, this is not a
randomized controlled trial and the number of subjects was small. Qureshi et al.
showed that palm vitamin E at 200 mg caused a reduction in serum total and LDL
cholesterol, apolipoprotein B, thromboxane, platelet factor 4 and glucose in subjects
during the four week-treatment [128]. A separate hypocholesterolemic group given
200 mg gamma-tocotrienol showed greater reduction in total cholesterol compared
to palm vitamin E group [128]. Daud et al. demonstrated that supplementation of
tocotrienol for 16 weeks (180 mg tocotrienols with 40 mg tocopherols) decreased
the normalized plasma TG and increased HDL in patients undergoing chronic
hemodialysis [36]. These changes might be associated with higher plasma
apolipoprotein and lower cholesteryl-ester transfer protein activity [36]. It should be
noted that the results of this study was not adjusted for medication used by the
subjects. Chin et al. showed that when elderly (>50 years) and young (<50 years)
subjects were supplemented with tocotrienol-rich fraction (160 mg/day, containing
74 % tocotrienols and 26 % tocopherols) for 6 months, the elderly subjects
responded better by showing an increase in HDL cholesterol compared to placebo
group [29]. The products of oxidation, such as protein carbonyl and
advance-glycation end products, were also reduced only in the elderly subjects [29].
Baliarsingh et al. supplemented 19 type 2 diabetic subjects with hyperlipidemia
with tocotrienol-rich fraction (3 mg/kg body weight) for 60 days and total lipids,
total and LDL cholesterol were reduced with treatment [11]. In the study by Qureshi
et al., hypercholesterolemic subjects on American Heart Association step 1 diet
were supplemented with rice bran tocotrienol-rich fraction at different doses (25–
200 mg/kg) [132]. They observed that 100 mg/kg tocotrienol-rich fraction pro-
duced maximum decrease in total and LDL cholesterol, apolipoprotein B and TG
compared to baseline [132]. In another study, Qureshi demonstrated that hyperc-
holesterolemic subjects on American Heart Society step 1 diet supplemented with
rice bran tocotrienol-rich fraction and lovastatin in combination for 25 weeks
showed more significant reduction than individual treatments [131]. The combi-
nation of alpha-tocopherol and lovastatin did not produce similar changes [131].
Tomeo et al. supplemented subjects presented with hyperlipidemia, cere-
brovascular disease and carotid stenosis with 300 mg palm vitamin E (16 mg
alpha-tocopherol and 40 mg tocotrienols in 240 mg palm olein) for 18 months
[169]. Apparent carotid atherosclerotic regression was found in supplemented
subjects while some controls showed progression [169]. Malondialdehyde was
decreased in the treatment group but was increased in the placebo group [169].
However, no changes in total and LDL cholesterol, TG and HDL were found in
both groups [169]. In men with mildly elevated serum lipid level, supplementation
of tocotrienol (4 capsules daily for 6 weeks, each containing 35 mg tocotrienols
and 20 mg alpha-tocopherol) produced no significant effects on serum LDL and
HDL cholesterol, TG, lipoprotein a, lipid peroxide, platelet aggregation velocity
and maximum aggregation, and thromboxane B2 formation compared to men
5 Tocotrienol and Its Role in Chronic Diseases 107
growth factor-1 beta and interleukin-1 beta in the sciatic nerve [78]. Apoptosis
indicated by caspase 3 expression was also decreased in the sciatic nerve [78].
There are limited studies attempted to validate the hypoglycemic effects of
tocotrienol in humans and the results are heterogenous. In a longitudinal study
(median follow-up period 10.2 years) assessing the intake of antioxidants and the
risk of type 2 diabetes in 29,133 Finnish male smokers aged 50–69 years,
Kataja-Tuomola et al. found that all tocotrienol isomers were not associated with
the risk of the disease after multiple adjustment. This could be contributed to the
low levels of tocotrienols in the subjects [71]. In the study of Baliarsingh et al., type
2 diabetic subjects supplemented with tocotrienol-rich fraction (3 mg/kg body
weight) for 60 days showed no changes in fasting and postprandial plasma sugar,
HbA1c and serum creatinine [11]. This could be due to the limited number of
subjects in this study (n = 19) and the low dose of tocotrienol. In a study by
Haghighat et al., type 2 diabetic subjects taking non-insulin medication were ran-
domized to take 15 mg/day tocotrienol mixture or blank canola oil with meals for
4 weeks [47]. It was observed that urinary microalbumin and serum high-sensitivity
C-reactive protein were significantly lowered in the tocotrienol group compared to
the placebo, indicating a better renal profile and a reduced risk for cardiovascular
disease [47].
activity of mature osteoclasts [46]. Brook et al. compared the suppressive effects of
different tocotrienol isomers (alpha, gamma and delta) on human-derived CD14+
cells [15]. It was found that gamma and delta-tocotrienol were effective in sup-
pressing the formation of osteoclast-like cells and the resorbing activity of these
cells was significantly inhibited. Gamma-tocotrienol was found to be with less toxic
to CD14+ cells, indicating that it is safer compared to delta-tocotrienol [15].
The effectiveness of tocotrienol in preventing bone loss has been demonstrated
in several animal models of osteoporosis. The methods of assessing bone health in
the animals included densitometry which measures bone mineral density; histo-
morphometry which measures bone structure, cells and mineralization activity;
biomechanical test which measures the stiffness and strength of the bone; bone
mineral concentration and serum bone remodelling markers which measure the
bone formation and resorption activity (reviewed in [24, 27]).
Norazlina supplemented ovariectomized rats with palm vitamin E, which was
rich in tocotrienol at either 30 or 60 mg/kg body weight for 8 months and measured
the femoral and vertebral bone mineral density [118]. It was found palm vitamin E
prevented the decline of bone mineral density in both supplemented groups [118].
Muhammad et al. observed that palm tocotrienol at 60 mg/kg body weight for
8 weeks could improve bone structural indices (increased bone volume and tra-
becular number, and decreased trabecular separation) in an oestrogen-deficient
model of osteoporosis induced by ovariectomy [96]. A decrease in osteoclast
number was found in the tocotrienol supplemented group [96]. The effects of
alpha-tocopherol at 60 mg/kg body weight were found to be comparable with palm
tocotrienol [96]. In a subsequent experiment, Muhammed et al. supplemented the
ovariectomized rats with tocotrienol-rich fraction (60 mg/kg for 8 weeks) and
found that it improved bone structural indices (increased bone volume and tra-
becular thickness, and decreased trabecular separation) [97]. Its effects were com-
parable to calcium (15 mg/kg) and oestrogen treatment (64.5 µg/kg) [97].
Aktifanus et al. demonstrated that tocotrienol-rich fraction at 60 mg/kg body weight
for 8 weeks improved bone dynamic indices (decreased single-labelled surface,
increased double-labelled surface, mineral apposition rate and bone formation rate)
in ovariectomized rats [8]. The effects of tocotrienol were better than oestrogen
replacement (64.5 µg/kg body weight) [8]. Using palm tocotrienol at 60 mg/kg
body weight for 8 weeks, Soelaiman et al. demonstrated similar findings and its
effects were superior to calcium supplementation (1 % calcium in drinking water
ad libitum) [154]. Abdul-Majeed et al. supplemented ovariectomized rats with
annatto tocotrienol (60 mg/kg body weight for 8 weeks) or in combination with
lovastatin (11 mg/kg body weight for 8 weeks) [3, 4]. Annatto tocotrienol alone
was found to improve bone structural (increased bone volume, trabecular number
and trabecular thickness, decreased trabecular separation) and cellular indices (in-
creased osteoblast number, osteoid surface and osteoid volume, and decreased
osteoclast number and eroded surface), bone biomechanical strength (increased
load, strain, stress and Young’s modulus) and bone remodelling markers (increased
osteocalcin, a bone formation marker and decreased type-1 C-terminal telopeptide
crosslink, a bone resorption marker) [3, 4]. The combination of annatto tocotrienol
112 K.-Y. Chin et al.
Anticancer activity of vitamin E was discovered and studied extensively since the
first experimental evidence of inhibition of tumorigenesis by vitamin E rich palm
oil [164]. Anticancer property of tocotrienol was found to be independent of its
antioxidant property [32, 162]. Tocotrienol isomer with lower antioxidant proper-
ties like delta- and gamma-tocotrienol usually showed greater potency in killing
cancerous cells. Based on quantitative structure–activity relationship studies,
chromanol structure and phytyl carbon tail were shown to be essential in apoptosis
induction [13]. Decreasing methyl groups on the chromanol head increased
anti-proliferative potency of tocotrienol (delta-tocotrienol > gamma-tocotrienol >
beta-tocotrienol > alpha-tocotrienol) [162]. Shortening the unsaturated phytyl tail
of gamma-tocotrienol by one isoprenyl unit also resulted in greater apoptotic
activity [13]. In silico analysis suggested that esterification of lysine with
gamma-tocotrienol could enhanced its anticancer activity [114]. Tocotrienol
metabolites such as carboxyethyl-hydroxychromans were found to be a less
effective anti-proliferative agent [34].
Besides that, tumor cell uptake and intracellular accumulation of tocotrienol also
determine its anticancer activity. Due to the lipophilic nature of tocotrienols, they
are readily transferred between the membranes and accumulated in cells in a time
and concentration-dependent manner [187]. Studies have shown that serum
physiological tocotrienol level was usually low or absent due to slow absorption,
rapid metabolism and lack of a specific carrier protein [164]. Besides that, toco-
trienol is highly tissue-dependent and it is only found in a few tissues/organs like
the liver and pancreas [52, 55]. Interestingly, in animal models, gamma-tocotrienol
and delta-tocotrienol were found to accumulate in tumors [52, 55]. Different
tocotrienol isomers possess different cellular accumulation tendency, i.e.
delta > gamma > alpha-tocotrienol [52, 87, 88, 116, 141, 144]. This might explain
the poor anticancer activity of alpha-tocotrienol.
Tocotrienol and tocotrienol-rich fraction were demonstrated to induce
anti-proliferation and apoptosis selectively in several cancerous cell lines origi-
nating from the mammary gland [32, 41, 44, 87, 109, 146, 173–175, 189], lung
[70, 86, 184], liver [49, 52, 140, 141, 172], prostate [33, 86, 158], melanoma
[22, 86], colon [40, 182] and leukemic cells [100]. The inhibitory potency of
tocotrienol-rich fraction, tocotrienol isoforms or their succinate synthetic deriva-
tives in different cancerous cells generally follows the order of delta- >gamma- and
beta- >alpha-tocotrienol [22, 33, 40, 52, 140, 172]. Studies also demonstrated that
tocotrienol possessed little or no adverse effect on normal cell viability or growth in
in vitro [49, 70, 87, 88, 141, 158, 184] or animal models [112, 135, 137]. In animal
studies, tocotrienols and tocotrienol-rich fraction were also demonstrated to reduce
tumor load. Oral administration of 0.05 % tocotrienol-rich vitamin E mixture sig-
nificantly reduced liver and lung carcinogenesis in C3H/He mice and 4NQO-treated
ddY mice model [172]. Gamma-tocotrienol or delta-tocotrienol diet (0.1 %) sig-
nificantly delayed tumor growth in C3H/HeN mice inoculated subcutaneously with
5 Tocotrienol and Its Role in Chronic Diseases 115
murine hepatoma MH134 cells [52]. There was no apparent adverse effect and
insignificant body and tissue weight changes in animals fed with 0.1 % of
gamma-tocotrienol or delta-tocotrienol diet [52].
Studies also demonstrated that tocotrienol as a farnesol-mimetic molecule and
HMGR inhibitor possessed great potential to produce synergistic anticancer activity
with other chemotherapeutic agents [86]. Combination of low concentration of
gamma-tocotrienol with several individual statins synergistically inhibited HMGR
activity and Rap1A & Rab6 prenylation [174]. Besides that, combination of
tocotrienol with flavonoids such as genistein, naringenin, hesperetin, tangeretin,
nobiletin, quercetin, apigenin, epigallocatechin gallate or resveratrol demonstrated
synergistic activity in inhibiting human breast cancer MCF7 or/and MDA-MB-435
cells proliferation, which was enhanced further with the addition of tamoxifen [45,
54]. The combination of tocotrienol with docetaxel also resulted in higher pro-
portion of apoptotic cells [22]. Combination of tocotrienols with tamoxifen
increased the arrest of DNA synthesis and triggered an endoplasmic reticulum
(ER)-independent anti-proliferation event in the breast cancer cells [44, 189].
Combination of tocotrienol isomers (delta-tocotrienol and gamma-tocotrienol) was
also found to induce cell death synergistically in DU145 and PC3 [33]. In animal
studies, gamma-tocotrienol also acted synergistically with lovastatin to reduce
tumor load in C57BL/6 mice inoculated with B16 cells [86].
There are limited clinical trials on tocotrienol and cancer conducted to date.
Malaysian Palm Oil Board had conducted two clinical trials on the efficacy of
tocotrienol in breast cancer patients [107]. The first study was conducted to identify
tocopherol and tocotrienol concentrations in malignant and benign adipose tissues
of breast cancer patients after a palm oil diet. The results revealed a higher con-
centration of tocotrienols (alpha-, delta- and gamma-isomers), in the adipose tissues
of patients with benign breast lumps compared with that of patients with malignant
tumors [107]. The second study was a 5-year double-blind, placebo-controlled pilot
trial conducted to compare tamoxifen with or without tocotrienol-rich fraction on
early breast cancer [108]. The risk of dying due to breast cancer decreased by 70 %
and risk of recurrence decreased by 20 % in patients taking the adjuvant tocotrienol
and tamoxifen compared with the patients receiving only tamoxifen [107].
However, there was no significant association between adjuvant tocotrienol therapy
with breast cancer survival rate [108].
Caspases are constitutively present in cells in an inactive precursor form, the pro-
caspases [164]. In receptor-mediated apoptosis, there is early activation of death
receptors (Fas, TNF or TRAIL) which lead to caspase-8 activation. In
mitochondria-mediated apoptosis, there are apoptotic signals from damaged DNA
116 K.-Y. Chin et al.
The inhibition of HMGR by tocotrienol not only suppresses cholesterol synthesis but
also inhibits cell proliferation via prenylation of Ras, RhoA or Rap1A signalling
molecules through generation of prenyl intermediates [184]. Farnesyl side chain of
tocotrienol is essential for the post-transcriptional suppression of HMGR.
Gamma-tocotrienol was shown to downregulate HMGR levels in +SA cells via
post-translational protein degradation [173]. However, the gamma-
tocotrienol-induced downregulation of HMGR was not able to block Rap1A &
Rab6 prenylation and trigger +SA cell death [174]. Delta-tocotrienol-induced growth
arrest might be due to HMGR inhibition, causing marked decrease of cyclin
D1/cyclin-dependent kinase CDK4 complex but not cyclin E/CDK2 complex, which
subsequently reduced the phosphorylation of retinoblastoma RB protein and E2F1
expression [41, 103].
6-O-carboxypropyl-alpha-tocotrienol (alpha-T3E) was more potent compared to
alpha-tocotrienol in inducing apoptosis in A549 cells and H28 cells [70, 184]. 6-O-
carboxypropyl-alpha-tocotrienol also harbours the similar farnesyl side chain and
might possess HMG-CoA reductase inhibitory activity. 6-O-carboxypropyl-
alpha-tocotrienol can inhibit RhoA geranyl-geranylation and Ras molecules farne-
sylation, subsequently blocking MEK, ERK and p38 phosphorylation and lead to
cyclin D and Bcl-xL downregulation during G1 arrest and apoptosis [160, 174, 184].
5 Tocotrienol and Its Role in Chronic Diseases 117
A toxicity study conducted by Ima-Nirwana et al. in the mice revealed that palm
tocotrienol at the doses of 500 and 1000 mg/kg body weight (oral) for 14 days
(subacute) and 42 days (subchronic) increased the bleeding the clotting time [57].
After conversion based on body surface area [138], this is equivalent to 40.54 and
81.08 mg/kg in humans. Nakamura et al. observed changes in haematology, blood
serum enzyme levels and organ histology in rats fed with palm tocotrienol
(0.75–3 % weight of diet) for 13 weeks [104]. The no-observed-adverse-effect level
derived from this study was 120 mg/kg body weight for male rats and 130 mg/kg
body weight for female rats [104]. This is equivalent to 19.46 mg/kg in men and
21.08 mg/kg in women. In view of these studies, it is recommended that patients
who are taking anticoagulants such as warfarin, heparin and aspirin to refrain from
using tocotrienol because it may increase haemorrhagic risk. Prolonged use of
tocotrienol at high dose should also be avoided because it may exert adverse effects
on the liver. Taking these into consideration, the use of tocotrienol is relatively safe.
The highest dose used in human recorded in this review is 400 mg [47], equivalent
to 5.71 mg/kg for a 70 kg adults, which is far below the toxic doses.
5.9 Conclusion
The two groups of vitamin E, the tocotrienols and the tocopherols possess distinct
similarities and differences. These differences may be seen even between isomers
within the same group. Extensive in vivo and in vitro studies on vitamin E mixtures
from various natural sources, as well as the pure isomers, have been done. Most of
these studies have shown beneficial effects of the tocotrienol and tocopherol iso-
mers in varying degrees. The source of the naturally occurring vitamin E mixtures
are from palm oil, rice bran or annatto oil. However, in the reported literature, the
vitamin E mixtures differ in composition, some consisting of mixtures of toco-
pherols and tocotrienols, while some are mixtures of tocotrienol isomers only. The
doses used also differ between studies. Even experiments using individual toco-
trienol or tocopherol isomers differ in their effective doses. Thus, it is not easy to
summarize the results, but in general the preclinical literature shows that toco-
trienols are beneficial in metabolic disease conditions associated with elevated free
radicals and inflammatory cytokines, such as dyslipidaemia, diabetes mellitus and
osteoporosis. Some clinical trials on tocotrienols and dyslipidaemia have been
reported, while none on diabetes mellitus or osteoporosis are available so far. Data
from the available clinical trials are inconclusive, and generally the beneficial
effects are slight and not as significantly obvious as the effects seen in the in vivo
and in vitro studies. This begs the question as to whether dose of the tocotrienols
used in the human studies were too low, or the duration of treatment insufficient, or
even whether the sampling methods were adequate. Another cause for the
5 Tocotrienol and Its Role in Chronic Diseases 121
discrepancy could be that the distribution and metabolism of the tocotrienols differ
between humans and the rodent models used. It will be interesting to see whether
the same sort of observations will be seen in clinical trials on the effects of
tocotrienols in diabetics or in osteoporotic patients.
Many well-reported studies on the anticancer effects of tocotrienol were found in
the literature. Most of these studies are in vitro studies using cancer cell lines. Most
of the studies are on breast cancer cells, while some studies are on liver, colon, lung
and leukaemic cell lines. The mechanisms have been elucidated as being apoptotic
and anti-angiogenetic. The anticancer effects of tocotrienol are thought to be sep-
arate from the antioxidant and anti-inflammatory effects. Limited animal studies
have been done using genetically modified mice, and only two reported clinical
trials on the effects of tocotrienols on breast cancer are found in literature. Again the
data from the human studies are scant and inconclusive.
From the above review, tocotrienol presents an exciting possibility as an alter-
native or adjuvant therapy for diseases associated with increased oxidative stress
and inflammation, such as dyslipidaemia and diabetes. Its application in preventing
osteoporosis in humans still awaits results from clinical trial. More coordinated,
well-designed clinical trials are needed to determine the effects on humans, how-
ever, the consistently beneficial effects in in vivo studies indicate that the potential
for human therapeutic use is real. As for the anticancer properties, the potential for
use presents a real challenge to researchers. The apoptotic and anti-angiogenetic
properties seen in the cell culture studies appears very promising and can be the
alternative to the stressful chemotherapy, or at least as an adjuvant to reduce the
side-effects of chemotherapy.
References
7. Ahmad NS, Khalid BAK, Luke DA, Ima Nirwana S (2005) Tocotrienol offers better
protection than tocopherol from free radical-induced damage of rat bone. Clin Exp
Pharmacol Physiol 32(9):761–770
8. Aktifanus AT, Shuid AN, Rashid NH et al (2012) Comparison of the effects of tocotrienol
and estrogen on the bone markers and dynamic changes in postmenopausal osteoporosis rat
model. Asian J Anim Vet Adv 7(3):225–234
9. Atli T, Keven K, Avci A et al (2004) oxidative stress and antioxidant status in elderly
diabetes mellitus and glucose intolerance patients. Arch Gerontol Geriatr 39(3):269–275
10. Baek KH, Oh KW, Lee WY et al (2010) Association of oxidative stress with postmenopausal
osteoporosis and the effects of hydrogen peroxide on osteoclast formation in human bone
marrow cell cultures. Calcif Tissue Int 87(3):226–235
11. Baliarsingh S, Beg ZH, Ahmad J (2005) The therapeutic impacts of tocotrienols in type 2
diabetic patients with hyperlipidemia. Atherosclerosis 182(2):367–374
12. Barnham KJ, Masters CL, Bush AI (2004) Neurodegenerative diseases and oxidative stress.
Nat Rev Drug Discov 3(3):205–214
13. Birringer M, EyTina JH, Salvatore BA, Neuzil J (2003) Vitamin E analogues as inducers of
apoptosis: structure-function relation. Br J Cancer 88(12):1948–1955
14. Brigelius-Flohe R, Kelly FJ, Salonen JT, Neuzil J, Zingg JM, Azzi A (2002) The European
perspective on vitamin E: current knowledge and future research. Am J Clin Nutr 76(4):703–
716
15. Brooks R, Kalia P, Ireland DC, Beeton C, Rushton N (2011) Direct inhibition of osteoclast
formation and activity by the vitamin E isomer gamma-tocotrienol. Int J Vitam Nutr Res 81
(6):358–367
16. Budin SB, Othman F, Louis SR, Bakar MA, Das S, Mohamed J (2009) The effects of palm
oil tocotrienol-rich fraction supplementation on biochemical parameters, oxidative stress and
the vascular wall of streptozotocin-induced diabetic rats. Clinics (Sao Paulo) 64(3):235–244
17. Buhaescu I, Izzedine H (2007) Mevalonate pathway: a review of clinical and therapeutical
implications. Clin Biochem 40(9–10):575–584
18. Burdeos GC, Nakagawa K, Kimura F, Miyazawa T (2012) Tocotrienol attenuates triglyceride
accumulation in Hepg2 cells and F344 rats. Lipids 47(5):471–481
19. Burdeos GC, Nakagawa K, Watanabe A, Kimura F, Miyazawa T (2013) Gamma-tocotrienol
attenuates triglyceride through effect on lipogenic gene expressions in mouse hepatocellular
carcinoma hepa 1-6. J Nutr Sci Vitaminol (Tokyo) 59(2):148–151
20. Callaway DA, Jiang JX (2015) Reactive oxygen species and oxidative stress in
osteoclastogenesis, skeletal aging and bone diseases. J Bone Miner Metab 33(4):359–370
21. Campbell PT, Newton CC, Patel AV, Jacobs EJ, Gapstur SM (2012) Diabetes and
cause-specific mortality in a prospective cohort of one million U.S. adults. Diabetes Care 35
(9):1835–1844
22. Chang PN, Yap WN, Lee DT, Ling MT, Wong YC, Yap YL (2009) Evidence of
Γ-tocotrienol as an apoptosis-inducing, invasion-suppressing, and chemotherapy
drug-sensitizing agent in human melanoma cells. Nutr Cancer 61(3):357–366
23. Chatelain E, Boscoboinik DO, Bartoli G-M et al (1993) Inhibition of smooth muscle cell
proliferation and protein kinase C activity by tocopherols and tocotrienols. Biochim Biophys
Acta (BBA) Mol Cell Res 1176(1–2):83–89
24. Chin K-Y, Ima-Nirwana S (2012) Vitamin E as an antiosteoporotic agent via receptor
activator of nuclear factor kappa-B ligand signaling disruption: current evidence and other
potential research areas. Evid Based Complement Altern Med 2012:747020
25. Chin KY, Abdul-Majeed S, Fozi NF, Ima-Nirwana S (2014) Annatto tocotrienol improves
indices of bone static histomorphometry in osteoporosis due to testosterone deficiency in rats.
Nutrients 6(11):4974–4983
26. Chin KY, Ima-Nirwana S (2014) The effects of alpha-tocopherol on bone: a double-edged
sword? Nutrients 6(4):1424–1441
27. Chin KY, Ima-Nirwana S (2015) the biological effects of tocotrienol on bone: a review on
evidence from rodent models. Drug Des Devel Ther 9:2049–2061
5 Tocotrienol and Its Role in Chronic Diseases 123
28. Chin KY, Ima Nirwana S (2014) The effects of annatto-derived tocotrienol supplementation
in osteoporosis induced by testosterone deficiency in rats. Clin Interv Aging 9:1247–1259
29. Chin SF, Ibahim J, Makpol S et al (2011) Tocotrienol rich fraction supplementation
improved lipid profile and oxidative status in healthy older adults: a randomized controlled
study. Nutr Metab (Lond) 8(1):42
30. Choy EHS, Panayi GS (2001) Cytokine pathways and joint inflammation in rheumatoid
arthritis. N Engl J Med 344(12):907–916
31. Chun J, Lee J, Ye L, Exler J, Eitenmiller RR (2006) Tocopherol and tocotrienol contents of
raw and processed fruits and vegetables in the United States Diet. J Food Compos Anal 19
(2–3):196–204
32. Comitato R, Nesaretnam K, Leoni G et al (2009) A novel mechanism of natural vitamin E
tocotrienol activity: involvement of Erβ signal transduction. Am J Physiol Endocrinol Metab
297:E427–E437
33. Constantinou C, Hyatt JA, Vraka PS et al (2009) Induction of caspase-independent
programmed cell death by vitamin E natural homologs and synthetic derivatives. Nutr Cancer
61(6):864–874
34. Conte C, Floridi A, Aisa C, Piroddi M, Floridi A, Galli F (2004) Γ-tocotrienol metabolism
and antiproliferative effect in prostate cancer cells. Ann N Y Acad Sci 1031:391–394
35. Dandona P, Aljada A, Bandyopadhyay A (2004) Inflammation: the link between insulin
resistance, obesity and diabetes. Trends Immunol 25(1):4–7
36. Daud ZA, Tubie B, Sheyman M et al (2013) Vitamin E tocotrienol supplementation
improves lipid profiles in chronic hemodialysis patients. Vasc Health Risk Manag 9:747–761
37. Deng L, Ding Y, Peng Y et al (2014) Γ-tocotrienol protects against ovariectomy-induced
bone loss via mevalonate pathway as Hmg-Coa reductase inhibitor. Bone 67:200–207
38. Deshpande AD, Harris-Hayes M, Schootman M (2008) Epidemiology of diabetes and
diabetes-related complications. Phys Ther 88(11):1254–1264
39. Di Marco E, Jha JC, Sharma A, Wilkinson-Berka JL, Jandeleit-Dahm KA, de Haan JB
(2015) Are reactive oxygen species still the basis for diabetic complications? Clin Sci (Lond)
129(2):199–216
40. Eitsuka T, Nakagawa K, Miyazawa T (2006) Down-regulation of telomerase activity in
Dld-1 human colorectal adenocarcinoma cells by tocotrienol. Biochem Biophys Res
Commun 348(1):170–175
41. Elangovan S, Hsieh TC, Wu JM (2008) Growth inhibition of human Mda-Mb-231 breast
cancer cells by Δ-tocotrienol is associated with loss of cyclin D1/Cdk4 expression and
accompanying changes in the state of phosphorylation of the retinoblastoma tumor
suppressor gene product. Anticancer Res 28:2641–2648
42. Frega N, Mozzon M, Bocci F (1998) Identification and estimation of tocotrienols in the
annatto lipid fraction by gas chromatography-mass spectrometry. J Am Oil Chem Soc 75
(12):1723–1727
43. Gee PT (2011) Unleashing the untold and misunderstood observations on vitamin E. Genes
Nutr 6(1):5–16
44. Guthrie N, Gapor A, Chambers AF, Carroll KK (1997) Inhibition of proliferation of estrogen
receptor-negative Mda-Mb-435 and -Positive Mcf-7 human breast cancer cells by palm oil
tocotrienols and tamoxifen, alone and in combination. J Nutr 127:544S–548S
45. Guthrie N, Gapor A, Chambers AF, Carroll KK (1997) Palm oil tocotrienols and plant
flavonoids act synergistically with each other and with tamoxifen in inhibiting proliferation
and growth of estrogen receptor-negative Mda-Mb-435 and -positive Mcf-7 human breast
cancer cells in culture. Asia Pac J Clin Nutrition 6(1):41–45
46. Ha H, Lee JH, Kim HN, Lee ZH (2011) Alpha-tocotrienol inhibits osteoclastic bone
resorption by suppressing rankl expression and signaling and bone resorbing activity.
Biochem Biophys Res Commun 406(4):546–551
47. Haghighat N, Vafa M, Eghtesadi S, Heidari I, Hosseini A, Rostami A (2014) The effects of
tocotrienols added to canola oil on microalbuminuria, inflammation, and nitrosative stress in
124 K.-Y. Chin et al.
patients with type 2 diabetes: a randomized, double-blind, placebo-controlled trial. Int J Prev
Med 5(5):617–623
48. Halliwell B (1994) Free radicals, antioxidants, and human disease: curiosity, cause, or
consequence? Lancet 344(8924):721–724
49. Har CH, Keong CK (2005) Effects of tocotrienols on cell viability and apoptosis in normal
murine liver cells (Bnl Cl.2) and liver cancer cells (Bnl 1me A.7r.1), in Vitro. Asia Pac J Clin
Nutr 14(4):374–380
50. Heng KS, Hejar AR, Johnson Stanslas J, Ooi CF, Loh SF (2015) Potential of mixed
tocotrienol supplementation to reduce cholesterol and cytokines level in adults with
metabolic syndrome. Malays J Nutr 21(2):231–243
51. Hermizi H, Faizah O, Ima-Nirwana S, Ahmad Nazrun S, Norazlina M (2009) Beneficial
effects of tocotrienol and tocopherol on bone histomorphometric parameters in
Sprague-Dawley male rats after nicotine cessation. Calcif Tissue Int 84(1):65–74
52. Hiura Y, Tachibana H, Arakawa R et al (2009) Specific accumulation of Γ- and
Δ-tocotrienols in tumor and their antitumor effect in vivo. J Nutr Biochem 20(8):607–613
53. Hosomi A, Arita M, Sato Y et al (1997) Affinity for alpha-tocopherol transfer protein as a
determinant of the biological activities of vitamin E analogs. FEBS Lett 409(1):105–108
54. Hsieh TC, Wu JM (1992) Suppression of cell proliferation and gene expression by
combinatorial synergy of Egcg, resveratrol and Γ-tocotrienol in estrogen receptor-positive
Mcf-7 breast cancer cells. Int J Oncol 33(4):851–859
55. Husain K, Francois RA, Hutchinson SZ et al (2009) Vitamin E Δ-tocotrienol levels in tumor
and pancreatic tissue of mice after oral administration. Pharmacology 83(3):157–163
56. Ima-Nirwana S, Fakhrurazi H (2002) Palm vitamin E protects bone against
dexamethasone-induced osteoporosis in male rats. Med J Malaysia 57(2):133–141
57. Ima-Nirwana S, Nurshazwani Y, Nazrun AS, Norliza M, Norazlina M (2011) Subacute and
subchronic toxicity studies of palm vitamin E in mice. J Pharmacol Toxicol 6:166–173
58. Ima Nirwana S, Kiftiah A, Zainal A, Norazlina M, Abd. Gapor M, Khalid BAK (2000) Palm
vitamin E prevents osteoporosis in orchidectomised growing male rats. Nat Prod Sci 6
(4):155–160
59. Ima Nirwana S, Suhaniza S (2004) Effects of tocopherols and tocotrienols on body
composition and bone calcium content in adrenalectomized rats replaced with
dexamethasone. J Med Food 7(1):45–51
60. Inokuchi H, Hirokane H, Tsuzuki T, Nakagawa K, Igarashi M, Miyazawa T (2003)
Anti-angiogenic activity of tocotrienol. Biosci Biotechnol Biochem 67(7):1623–1627
61. Institute of Medicine (2000) Dietary reference intakes for vitamin C, vitamin E, selenium,
and carotenoids. The National Academies Press, Washington
62. Iqbal J, Minhajuddin M, Beg ZH (2003) Suppression of 7,12-dimethylbenz[alpha]
anthracene-induced carcinogenesis and hypercholesterolaemia in rats by tocotrienol-rich
fraction isolated from rice bran oil. Eur J Cancer Prev 12(6):447–453
63. Jiang Q (2014) Natural forms of vitamin E: metabolism, antioxidant, and anti-inflammatory
activities and their role in disease prevention and therapy. Free Radic Biol Med 72:76–90
64. Johnell O, Kanis J (2006) An estimate of the worldwide prevalence and disability associated
with osteoporotic fractures. Osteoporos Int 17(12):1726–1733
65. Jones MP, Pandak WM, Heuman DM, Chiang JY, Hylemon PB, Vlahcevic ZR (1993)
Cholesterol 7 alpha-hydroxylase: evidence for transcriptional regulation by cholesterol or
metabolic products of cholesterol in the rat. J Lipid Res 34(6):885–892
66. Kamat JP, Devasagayam TP (1995) Tocotrienols from palm oil as potent inhibitors of lipid
peroxidation and protein oxidation in rat brain mitochondria. Neurosci Lett 195(3):179–182
67. Kamat JP, Sarma HD, Devasagayam TP, Nesaretnam K, Basiron Y (1997) Tocotrienols from
palm oil as effective inhibitors of protein oxidation and lipid peroxidation in rat liver
microsomes. Mol Cell Biochem 170(1–2):131–137
68. Kanis JA, McCloskey EV, Harvey NC, Johansson H, Leslie WD (2015) Intervention
thresholds and the diagnosis of osteoporosis. J Bone Miner Res 30(10):1747–1753
5 Tocotrienol and Its Role in Chronic Diseases 125
function in men with mildly elevated serum lipid concentrations. Am J Clin Nutr 69(2):213–
219
91. Minhajuddin M, Beg ZH, Iqbal J (2005) Hypolipidemic and antioxidant properties of
tocotrienol rich fraction isolated from rice bran oil in experimentally induced hyperlipidemic
rats. Food Chem Toxicol 43(5):747–753
92. Miyazawa T, Inokuchi H, Hirokane H, Tsuzuki T, Nakagawa K, Igarashi M (2004)
Anti-angiogenic potential of tocotrienol in vitro. Biochemistry (Moscow) 69(1):67–69
93. Miyazawa T, Shibata A, Nakagawa K, Tsuzuki T (2008) Anti-angiogenic function of
tocotrienol. Asia Pac J Clin Nutr 17(S1):253–256
94. Miyazawa T, Tsuzuki T, Nakagawa K, Igarashi M (2004) Antiangiogenic potency of vitamin
E. Ann N Y Acad Sci 1031:401–404
95. Mizushina Y, Nakagawa K, Shibata A et al (2006) Inhibitory effect of tocotrienol on
eukaryotic DNA polymerase Λ and angiogenesis. Biochem Biophys Res Commun 339
(3):949–955
96. Muhammad N, Luke DA, Shuid AN, Mohamed N, Soelaiman IN (2012) Two different
isomers of vitamin E prevent bone loss in postmenopausal osteoporosis rat model. Evid
Based Complement Altern Med 2012:161527
97. Muhammad N, Razali S, Shuid AN, Mohamed N, Soelaiman IN (2013) Membandingkan
Kesan Antara Fraksi-Kaya Tokotrienol, Kalsium Dan Estrogen Terhadap Metabolisme
Tulang Tikus Terovariektomi. Sains Malays 42(11):1591–1597
98. Müller M, Cela J, Asensi-Fabado MA, Munné-Bosch S (2012) Tocotrienols in plants:
occurrence, biosynthesis, and function. In: Tan B, Watson RR, Preedy VR
(eds) Tocotrienols: vitamin E beyond tocopherols. CRC Press, Florida, pp 1–16
99. Mundy GR (2007) Osteoporosis and inflammation. Nutr Rev 65(suppl 3):S147–S151
100. Munteanu A, Ricciarelli R, Massone S, Zingg JM (2007) Modulation of proteasome activity
by vitamin E in Thp-1 monocytes. IUBMB Life 59(12):771–780
101. Mustad VA, Smith CA, Ruey PP, Edens NK, DeMichele SJ (2002) Supplementation with 3
compositionally different tocotrienol supplements does not improve cardiovascular disease
risk factors in men and women with hypercholesterolemia. Am J Clin Nutr 76(6):1237–1243
102. Naito Y, Shimozawa M, Kuroda M et al (2005) Tocotrienols reduce
25-hydroxycholesterol-induced monocyte-endothelial cell interaction by inhibiting the
surface expression of adhesion molecules. Atherosclerosis 180(1):19–25
103. Nakagawa K, Shibata A, Yamashita S et al (2007) In vivo angiogenesis is suppressed by
unsaturated vitamin E, tocotrienol. J Nutr 137:1938–1943
104. Nakamura H, Furukawa F, Nishikawa A et al (2001) Oral toxicity of a tocotrienol preparation
in rats. Food Chem Toxicol 39(8):799–805
105. Nazrun AS, Luke DA, Khalid BAK, Ima Nirwana S (2005) Vitamin E protects from
free-radical damage on femur of rats treated with ferric nitrilotriacetate. Curr Topics
Pharmacol 9(2):107–115
106. Nesaretnam K, Devasagayam TP, Singh BB, Basiron Y (1993) Influence of palm oil or its
tocotrienol-rich fraction on the lipid peroxidation potential of rat liver mitochondria and
microsomes. Biochem Mol Biol Int 30(1):159–167
107. Nesaretnam K, Meganathan P, Veerasenan SD, Selvaduray KR (2012) Tocotrienols and
breast cancer: the evidence to date. Genes Nutr 7(1):3–9
108. Nesaretnam K, Selvaduray KR, Abdul Razak G, Veerasenan SD, Gomez PA (2010)
Effectiveness of tocotrienol-rich fraction combined with tamoxifen in the management of
women with early breast cancer: a pilot clinical trial. Breast Cancer Res 12(5):R81
109. Nesaretnam K, Stephen R, Dils R, Darbre P (1998) Tocotrienols inhibit the growth of human
breast cancer cells irrespective of estrogen receptor status. Lipids 33(5):461–469
110. Ng LT, Ko HJ (2012) Comparative effects of tocotrienol-rich fraction, alpha-tocopherol and
alpha-tocopheryl acetate on inflammatory mediators and nuclear factor kappa B expression in
mouse peritoneal macrophages. Food Chem 134(2):920–925
111. Ng MH, Choo YM, Ma AN, Chuah CH, Hashim MA (2004) Separation of vitamin E
(tocopherol, tocotrienol, and tocomonoenol) in palm oil. Lipids 39(10):1031–1035
5 Tocotrienol and Its Role in Chronic Diseases 127
131. Qureshi AA, Sami SA, Salser WA, Khan FA (2001) Synergistic effect of tocotrienol-rich
fraction (Trf(25)) of rice bran and lovastatin on lipid parameters in hypercholesterolemic
humans. J Nutr Biochem 12(6):318–329
132. Qureshi AA, Sami SA, Salser WA, Khan FA (2002) Dose-dependent suppression of serum
cholesterol by tocotrienol-rich fraction (Trf25) of rice bran in hypercholesterolemic humans.
Atherosclerosis 161(1):199–207
133. Qureshi AA, Tan X, Reis JC et al (2011) Inhibition of nitric oxide in Lps-stimulated
macrophages of young and senescent mice by delta-tocotrienol and quercetin. Lipids Health
Dis 10:239
134. Raederstorff D, Elste V, Aebischer C, Weber P (2002) Effect of either gamma-tocotrienol or a
tocotrienol mixture on the plasma lipid profile in hamsters. Ann Nutr Metab 46(1):17–23
135. Rahmat A, Ngah WZW, Top AGM, Khalid BAK (1993) Long term tocotrienol
supplementation and glutathione-dependent enzymes during hepatocarcinogenesis in the
rat. Asia Pac J Clin Nutr 2:129–134
136. Rasool AH, Yuen KH, Yusoff K, Wong AR, Rahman AR (2006) Dose dependent elevation
of plasma tocotrienol levels and its effect on arterial compliance, plasma total antioxidant
status, and lipid profile in healthy humans supplemented with tocotrienol rich vitamin E.
J Nutr Sci Vitaminol (Tokyo) 52(6):473–478
137. Rasool AHG, Yuen KH, Yusoff K, Wong AR, Rahman ARA (2006) Dose dependent
elevation of plasma tocotrienol levels and its effect on arterial compliance, plasma total
antioxidant status, and lipid profile in healthy humans supplemented with tocotrienol rich
vitamin E. J Nutr Sci Vitaminol 52:473–478
138. Reagan-Shaw S, Nihal M, Ahmad N (2008) Dose translation from animal to human studies
revisited. FASEB J 22(3):659–661
139. Rickmann M, Vaquero EC, Malagelada JR, Molero X (2007) Tocotrienols induce apoptosis
and autophagy in rat pancreatic stellate cells through the mitochondrial death pathway.
Gastroenterology 132(7):2518–2532
140. Sakai M, Okabe M, Tachibana H, Yamada K (2006) Apoptosis induction by Γ-tocotrienol in
human hepatoma Hep3b cells. J Nutr Biochem 17(10):672–676
141. Sakai M, Okabe M, Yamasaki A, Tachibana H, Yamada K (2004) Induction of apoptosis by
tocotrienol in rat hepatoma Drlh-84 cells. Anticancer Res 24:1683–1688
142. Salman Khan M, Akhtar S, Al-Sagair OA, Arif JM (2011) Protective effect of dietary
tocotrienols against infection and inflammation-induced hyperlipidemia: an in vivo and in
silico study. Phytother Res PTR 25(11):1586–1595
143. Samant GV, Sylvester PW (2006) Γ-tocotrienol inhibits Erbb3-dependent Pi3k-Akt
mitogenic signalling in neoplastic mammary epithelial cells. Cell Prolif 39:563–574
144. Sen CK, Khanna S, Roy S, Packer L (2000) Molecular basis of vitamin E action: tocotrienol
potently inhibits glutamate-induced Pp60c-Src kinase activation and death of Ht4 neuronal
cells. J Biol Chem 275(17):13049–13055
145. Serbinova E, Kagan V, Han D, Packer L (1991) Free radical recycling and intramembrane
mobility in the antioxidant properties of alpha-tocopherol and alpha-tocotrienol. Free Radic
Biol Med 10(5):263–275
146. Shah SJ, Sylvester PW (2005) Tocotrienol-induced cytotoxicity is unrelated to mitochondrial
stress apoptotic signaling in neoplastic mammary epithelial cells. Biochem Cell Biol 83
(1):86–95
147. Shibata A, Nakagawa K, Shirakawa H et al (2012) Physiological effects and tissue
distribution from large doses of tocotrienol in rats. Biosci Biotechnol Biochem 76(9):1805–
1808
148. Shibata A, Nakagawa K, Sookwong P et al (2008) Tocotrienol inhibits secretion of
angiogenic factors from human colorectal adenocarcinoma cells by suppressing
hypoxia-inducible factor-1α. J Nutr 138(11):2136–2142
149. Shuid A, Mehat Z, Mohamed N, Muhammad N, Soelaiman I (2010) Vitamin E exhibits bone
anabolic actions in normal male rats. J Bone Miner Metab 28(2):149–156
5 Tocotrienol and Its Role in Chronic Diseases 129
150. Siddiqui S, Ahsan H, Khan MR, Siddiqui WA (2013) Protective effects of tocotrienols
against lipid-induced nephropathy in experimental type-2 diabetic rats by modulation in
Tgf-beta expression. Toxicol Appl Pharmacol 273(2):314–324
151. Siddiqui S, Rashid Khan M, Siddiqui WA (2010) Comparative hypoglycemic and
nephroprotective effects of tocotrienol rich fraction (Trf) from palm oil and rice bran oil
against hyperglycemia induced nephropathy in type 1 diabetic rats. Chem Biol Interact 188
(3):651–658
152. Sies H (1997) Oxidative stress: oxidants and antioxidants. Exp Physiol 82(2):291–295
153. Sies H (2000) What is oxidative stress? In: Keaney J, Jr. (ed) Oxidative stress and vascular
disease. Developments in cardiovascular medicine, vol 224. Springer, New York , pp 1–8
154. Soelaiman IN, Ming W, Abu Bakar R et al (2012) Palm tocotrienol supplementation
enhanced bone formation in oestrogen-deficient rats. Int J Endocrinol 2012:532862
155. Song BL, DeBose-Boyd RA (2006) Insig-dependent ubiquitination and degradation of
3-hydroxy-3-methylglutaryl coenzyme a reductase stimulated by delta- and
gamma-tocotrienols. J Biol Chem 281(35):25054–25061
156. Sookwong P, Nakagawa K, Murata K, Kojima Y, Miyazawa T (2007) Quantitation of
tocotrienol and tocopherol in various rice brans. J Agric Food Chem 55(2):461–466
157. Sookwong P, Nakagawa K, Yamaguchi Y et al (2010) Tocotrienol distribution in foods:
estimation of daily tocotrienol intake of Japanese population. J Agric Food Chem 58
(6):3350–3355
158. Srivastava JK, Gupta S (2006) Tocotrienol-rich fraction of palm oil induces cell cycle arrest
and apoptosis selectively in human prostate cancer cells. Biochem Biophys Res Commun
346(2):447–453
159. Stamler J, Daviglus ML, Garside DB, Dyer AR, Greenland P, Neaton JD (2000) Relationship
of baseline serum cholesterol levels in 3 large cohorts of younger men to long-term coronary,
cardiovascular, and all-cause mortality and to longevity. JAMA 284(3):311–318
160. Sun W, Xu W, Liu H et al (2009) Γ-tocotrienol induces mitochondria-mediated apoptosis in
human gastric adenocarcinoma Sgc-7901 cells. J Nutr Biochem 20(4):276–284
161. Suzuki YJ, Tsuchiya M, Wassall SR et al (1993) Structural and dynamic membrane
properties of alpha-tocopherol and alpha-tocotrienol: implication to the molecular mechanism
of their antioxidant potency. Biochemistry 32(40):10692–10699
162. Sylvester PW, Nachnani A, Shah S, Briski KP (2002) Role of Gtp-binding proteins in
reversing the antiproliferative effects of tocotrienols in preneoplastic mammary epithelial
cells. Asia Pac J Clin Nutr 11(Suppl):S452–S459
163. Sylvester PW, Shah S (2005) Intracellular mechanisms mediating tocotrienol-induced
apoptosis in neoplastic mammary epithelial cells. Asia Pac J Clin Nutr 14(4):366–373
164. Sylvester PW, Shah SJ (2005) Mechanisms mediating the antiproliferative and apoptotic
effects of vitamin E in mammary cancer cells. Front Biosci 10(1–3):699
165. Takahashi K, Loo G (2004) Disruption of mitochondria during tocotrienol-induced apoptosis
in Mda-Mb-231 human breast cancer cells. Biochem Pharmacol 67(2):315–324
166. Tan CY, Saw TY, Fong CW, Ho HK (2015) Comparative hepatoprotective effects of
tocotrienol analogs against drug-induced liver injury. Redox Biol 4:308–320
167. Tan DT, Khor HT, Low WH, Ali A, Gapor A (1991) Effect of a palm-oil-vitamin E
concentrate on the serum and lipoprotein lipids in humans. Am J Clin Nutr 53(4
Suppl):1027s–1030s
168. Theriault A, Wang Q, Gapor A, Adeli K (1999) Effects of gamma-tocotrienol on apob
synthesis, degradation, and secretion in Hepg2 cells. Arterioscler Thromb Vasc Biol 19
(3):704–712
169. Tomeo AC, Geller M, Watkins TR, Gapor A, Bierenbaum ML (1995) Antioxidant effects of
tocotrienols in patients with hyperlipidemia and carotid stenosis. Lipids 30(12):1179–1183
170. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J (2007) Free radicals and
antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol
39(1):44–84
130 K.-Y. Chin et al.
171. Vester H, Holzer N, Neumaier M, Lilianna S, Nussler AK, Seeliger C (2014) Green tea
extract (Gte) improves differentiation in human osteoblasts during oxidative stress. J Inflamm
(Lond) 11:15
172. Wada S, Satomi Y, Murakoshi M, Noguchi N, Yoshikawa T, Nishino H (2005) Tumor
suppressive effects of tocotrienol in vivo and in vitro. Cancer Lett 229(2):181–191
173. Wali VB, Bachawal SV, Sylvester PW (2009) Endoplasmic reticulum stress mediates
Γ-tocotrienol-induced apoptosis in mammary tumor cells. Apoptosis 14(11):1366–1377
174. Wali VB, Bachawal SV, Sylvester PW (2009) Suppression in mevalonate synthesis mediates
antitumor effects of combined statin and Γ-tocotrienol treatment. Lipids 44(10):925–934
175. Wali VB, Sylvester PW (2007) Synergistic antiproliferative effects of Γ-tocotrienol and statin
treatment on mammary tumor cells. Lipids 42(12):1113–1123
176. Wan Nazaimoon WM, Khalid BA (2002) Tocotrienols-rich diet decreases advanced
glycosylation end-products in non-diabetic rats and improves glycemic control in
streptozotocin-induced diabetic rats. Malays J Pathol 24(2):77–82
177. Wang Y, Jiang Q (2013) Gamma-tocotrienol inhibits lipopolysaccharide-induced
interlukin-6 and granulocyte colony-stimulating factor by suppressing C/Ebpbeta and
Nf-Kappab in macrophages. J Nutr Biochem 24(6):1146–1152
178. Wang Y, Park NY, Jang Y, Ma A, Jiang Q (2015) Vitamin E gamma-tocotrienol inhibits
cytokine-stimulated Nf-kappab activation by induction of anti-inflammatory A20 via stress
adaptive response due to modulation of sphingolipids. J Immunol 195(1):126–133
179. Watkins T, Lenz P, Gapor A, Struck M, Tomeo A, Bierenbaum M (1993)
Gamma-tocotrienol as a hypocholesterolemic and antioxidant agent in rats fed atherogenic
diets. Lipids 28(12):1113–1118
180. Weng-Yew W, Selvaduray KR, Ming CH, Nesaretnam K (2009) Suppression of tumor
growth by palm tocotrienols via the attenuation of angiogenesis. Nutr Cancer 61(3):367–373
181. Wu SJ, Liu PL, Ng LT (2008) Tocotrienol-rich fraction of palm oil exhibits
anti-inflammatory property by suppressing the expression of inflammatory mediators in
human monocytic cells. Mol Nutr Food Res 52(8):921–929
182. Xu WL, Liu JR, Liu HK et al (2009) Inhibition of proliferation and induction of apoptosis by
Γ-tocotrienol in human colon carcinoma Ht-29 cells. Nutrition 25(5):555–566
183. Yam ML, Abdul Hafid SR, Cheng HM, Nesaretnam K (2009) Tocotrienols suppress
proinflammatory markers and cyclooxygenase-2 expression in Raw264.7 macrophages.
Lipids 44(9):787–789
184. Yano Y, Satoh H, Fukumoto K et al (2005) Induction of cytotoxicity in human lung
adenocarcinoma cells by 6-O-carboxypropyl-Α-tocotrienol, a redox-silent derivative of
Α-tocotrienol. Int J Cancer 115(5):839–846
185. Yap SP, Yuen KH, Wong JW (2001) Pharmacokinetics and bioavailability of alpha-,
gamma- and delta-tocotrienols under different food status. J Pharm Pharmacol 53(1):67–71
186. Yoshida Y, Hayakawa M, Habuchi Y, Itoh N, Niki E (2007) Evaluation of lipophilic
antioxidant efficacy in vivo by the biomarkers hydroxyoctadecadienoic acid and isoprostane.
Lipids 42(5):463–472
187. Yoshida Y, Niki E, Noguchi N (2003) Comparative study on the action of tocopherols and
tocotrienols as antioxidant: chemical and physical effects. Chem Phys Lipids 123(1):63–75
188. Yu SG, Thomas AM, Gapor A, Tan B, Qureshi N, Qureshi AA (2006) Dose-response impact
of various tocotrienols on serum lipid parameters in 5-week-old female chickens. Lipids 41
(5):453–461
189. Yu W, Simmons-Menchaca M, Gapor A, Sanders BG, Kline K (1999) Induction of apoptosis
in human breast cancer cells by tocopherols and tocotrienols. Nutr Cancer 33(1):26–32
190. Zaiden N, Yap WN, Ong S et al (2010) Gamma delta tocotrienols reduce hepatic triglyceride
synthesis and Vldl secretion. J Atheroscler Thromb 17(10):1019–1032
Chapter 6
Indole-3-Carbinol and Its Role
in Chronic Diseases
Keywords Indole-3-carbinol DIM Signaling pathways Chronic diseases
Animal models Dietary intervention trials
6.1 Introduction
The plant family Cruciferae, particularly members of the genus Brassica, like
cabbage, broccoli, cauliflower, Brussels sprouts, kale, bok choy are rich sources of
sulfur-containing glucosinolates. These secondary products of plant metabolism
include, among others, glucobrassicin and neoglucobrassicin. When plant tissue is
disrupted, an endogenous thioglucosidase (myrosinase) is activated and converts
intestines, and liver after 1 h following oral administration of I3C to rats [39]. In
human volunteers intervention trial DIM was detected in plasma within 8 h fol-
lowing of 400 mg I3C oral dose [9]. In a phase I clinical trial in women, no I3C was
found in the plasma after administration of a single dose of up to 1200 mg or
multiple-doses at 400 mg provided twice daily for 4 weeks, and DIM was the only
detectable I3C-derived compound in plasma [105]. Fujioka et al. [47] have found
that urinary DIM level after uptake of I3C from Brussels sprouts or cabbage is a
biomarker of glucobrassicin exposure in humans. All these results support the
suggestion that I3C serves as the prodrug rather than the therapeutic agent itself. In
this regard, purified I3C as treatment agent used in in vitro models seems to be
somewhat contradictory, because there is no certainty that any metabolism of DIM
in cells occur. Thus, in this chapter the biological activity and the role in chronic
diseases will refer not only to I3C but also to DIM, its major condensation product
in humans.
I3C affects multiple signaling pathways and target molecules controlling cell
division, apoptosis or angiogenesis deregulated in cancer cells. Figure 6.2 presents
the overview of the signaling pathways and possible crosstalks influenced by I3C or
DIM. One of the major pathways targeted by I3C is phosphoinositide 3-kinase
134 B. Licznerska and W. Baer-Dubowska
membrane
Wnt
c ell
PI3K β -catenin
Reversal of MDR
GSK-3 β
Akt
MAPK
mTOR AUTOPHAGY
ANGIOGENESIS
NF κ B ERα
AR
nucleus
ERβ AhR
Sp1
Nrf2 BRCA1
miR -21
miR -146 DNA REPAIR
p27
Fig. 6.2 Signaling pathways and proposed crosstalks ($) affected by I3C/DIM; red-inhibition,
green-induction (AhR-aryl hydrocarbon receptor, AKR-aldo-keto reductase, Akt- protein kinase
B, AR-androgen receptor, BRCA-breast cancer tumor suppressor gene, CDK-cyclin-dependent
kinase, COX-cyclooxygenase, EGFR-epidermal growth factor receptor, ER-estrogen receptor,
GJIC-Gap junctional intracellular communication, GSK-glycogen synthase kinase,
GST-glutathione-S-transferases, HER-receptor tyrosine-protein kinase erbB-2,
MAPK-mitogen-activated protein kinases, MDR-multidrug resistance, NFjB-nuclear factor
kappa-light-chain-enhancer of activated B cells, Nrf2-nuclear transcription factor 2, NQO-NAD
(P)H:quinone oxidoreductases, PI3K-phosphoinositide 3-kinase, Sp1-Sp1 transcription factor)
and prostate cancer cell lines, human colon cancer xenografts in nude mice and in
mouse prostate cells, influencing apoptotic proteins, like p21, p27, and involved in
inflammation COX-2 [46].
I3C and DIM were found to downregulate miR-21 and miRNA-146, respec-
tively, in Panc-1 pancreatic cancer cells, which was related to induction of
chemosensitivity to gemcitabine in these cells [78, 98]. In another study, I3C
reversed the upregulation of miR-21 caused by lung carcinogen—vinyl carbamate
in mice [88]. DIM upregulated miR-21 in breast cancer MCF7 cells resulting in
reduced proliferation [62], and the let-7 family miRNA leading to inhibition of
self-renewal and clonogenic capacity of prostate cancer cells [69, 70].
To sum up, both I3C and its dimer demonstrate pleiotropic effects on cell sig-
naling and subsequently gene expression regulation. Some of these activities are
summarized in Table 6.2.
Table 6.2 Summary of the major biological effects of indole-3-carbinol (I3C) and its
condensation product—3,3’-diindolylmethane (DIM)
Effecta Target molecules I3C DIM
In In In In
vitro vivo vitro vivo
Induction of phase I CYP1A1, CYP1A2, + + +
enzymes CYP1B1
Induction of phase II GST, NQO + + + +
enzymes
Inhibition of DNA CYP1B1 +
adducts formation
Anti-estrogenic activity ER, AhR + + +
Anti-androgenic activity AR + +
Cell cycle arrest p21, p27, CDK6 + +
Pro-apoptotic Akt, NFjB, GSK3b, JNK + +
Anti-angiogenic activity E-cadherin, a-, b-, and + + +
c-catenins, MMP9
Anti-proliferative activityERb/ERa + + +
DNA repair BRCA1, RAD51 + + +
Reversal of MDR P-glycoprotein + +
Epigenetic modifications DNMTs, HDACs, miRNAs + +
Anti-inflammatory NFjB, COX-2 + + +
activity
AhR aryl hydrocarbon receptor, Akt protein kinase B, AR androgen receptor, BRCA breast cancer
tumor suppressor gene, CDK cyclin-dependent kinase, DNMT DNA methyltransferase,
ER-estrogen receptor, GSK glycogen synthase kinase, GST glutathione-S-transferases, HDAC
histone deacetylases, JNK c-Jun N-terminal kinase, MDR multidrug resistance, MMP matrix
metalloproteinase, NFjB nuclear factor kappa-light-chain-enhancer of activated B cells, NQO
NAD(P)H:quinone oxidoreductases
a
References in the text
138 B. Licznerska and W. Baer-Dubowska
The term “chronic diseases” appear under different names in different contexts.
According to WHO this term suggests the following shared features:
• The chronic disease epidemics take decades to become fully established—they
have their origins at young age;
• Given their long duration, there are many opportunities for prevention;
• They require a long-term and systematic approach to treatment (WHO Report
2015).
In this context, cardiovascular diseases, chronic respiratory diseases, diabetes
along with cancer are mentioned. However, chronic character of cancer is not so
obvious as in the case of the other illnesses. The idea of considering cancer as
chronic disease emerged recently when it was noted that many cancers, while still
very serious could be manageable chronic diseases with ongoing surveillance
and/or treatment. While this vision has not yet become a reality for most forms of
cancer, the past 10–20 years have brought about a marked acceleration in advance
toward this goal. In this regard some types of metastatic breast cancer have become
manageable over the long term, perhaps most famously with tamoxifen, which can
slow or stop malignant cell growth in many women with estrogen-dependent cancer
by blocking hormone receptors on tumor cells. Moreover, a new class of aromatase
(CYP19) inhibitors that target estrogen production was developed, which seem to
provide better results than tamoxifen [127]. I3C can also inhibit the expression of
aromatase as well as CYP isoforms involved in estrogen metabolism. Therefore,
I3C, as well as its condensation product DIM, appear to be a promising agent for
the prevention or recurrence of human tumors, particularly hormone-dependent
cancers. There are also data suggesting that I3C or DIM might also be useful for
prevention or recurrence of cardiovascular diseases, diabetes, or recurrent respira-
tory papillomatosis, as well as obesity.
It is almost 40 years after Lee Wattenberg and William Loub [122] for the first time
reported that I3C inhibited chemically induced breast and forestomach neoplasia in
rodents. Since then the antitumor activity of dietary I3C has been widely studied
and the inhibition of the development of other cancer types, including liver [95],
lung [63], and prostate [110] have been demonstrated.
Breast cancer is the most common and the leading cause of cancer mortality
among women worldwide. The preventive efficacy of I3C on mammary carcinoma
first observed in animal models, was confirmed in many mechanistic studies in cell
cultures and was supported by epidemiological studies with cruciferous vegetables
and their extracts or juices [117, 120].
6 Indole-3-Carbinol and Its Role in Chronic Diseases 139
One of the most important risk factor of breast tumors are estrogens, which are
classified as carcinogenic in humans [58]. These steroid hormones may contribute
to breast cancer development in two ways: (i) acting as promoters by stimulating
cell proliferation, (ii) inducing genotoxicity through the reaction of their active
metabolites with DNA thus acting as tumor initiators.
Experimental studies in vitro in breast epithelial cell lines showed that I3C, DIM,
and cabbage juices induce CYP450 genes CYP1A1, 1A2, 1B1 encoding the key
enzymes of estrogen catabolism. The profile of metabolites was in favor to
2-hydroxyestrogens being noncarcinogenic in comparison to estradiol and
4-hydroxy derivatives [80, 114]. This anti-estrogenic activity of I3C could be
explained by the induction of AhR receptor showed in other studies [114].
Moreover, I3C, DIM, and cabbage juices are capable of upregulating phase II
detoxifying enzymes, including GST and NQO in breast cancer cell lines [114,
120]. Upregulation of GSTs and NQO1 by I3C was correlated with increased levels
of Nrf2, in benign MCF7 and aggressive MDA-MB-231 breast cancer cell lines.
Thus, it may be assumed that I3C protects against estrogen-associated carcino-
genesis by removal of the genotoxic metabolites of estrogens.
Simultaneously, I3C and DIM influenced in situ production of estrogens in
breast epithelial MCF7 cancer cells by reducing the expression of aromatase
(CYP19), the enzyme that synthesizes estrogens by converting C19 androgens into
aromatic C18 estrogenic steroids [80]. Several studies have shown that there is an
overexpression of aromatase gene in breast cancer tissue [82]. Interestingly, the
potential of I3C to reduce estrogenic activity in the breast cancer cells was con-
firmed by other mechanisms, particularly via decreasing the AKR1C3 expression
mentioned in the previous section. In the mammary gland where enzyme converts
androstenedione to testosterone––one of the aromatase substrates [92, 101].
Besides the interference with estrogens metabolism pathways, I3C also affects
DNA repair, the cell cycle progression and apoptosis in breast cancer cell lines. In
this regard it was shown that I3C induces BRCA1 expression and that both I3C and
BRCA1 inhibited estrogen (E2)-stimulated ERa activity in human breast cancer
cells [44]. BRCA1 is DNA repair factor involved in repair of DNA double-strand
breaks. BRCA1gene expression is reduced or completely silenced in a significant
proportion of sporadic breast cancer because of hypermethylation of the gene
promoter [106, 126]. Thus, I3C-induced BRCA1 expression and inhibition of
estrogen-stimulated ERa activity by I3C and BRCA1 showed by some studies
could be one of the antitumor activity of the indole [44].
Several lines of evidence suggest I3C ability to arrest cell cycle in breast cancer
cells. In this regard I3C was reported to inhibit CDK2 activity in breast cancer
MCF7 cell line [48]. Moreover, both I3C and DIM upregulated CDK inhibitors p21
and p27, although in a very high concentration of 200 µM. Activity of this protein
is especially critical during the G1 to S phase transition. Consequently, the ability to
arrest G1 phase by I3C was shown [30]. Other studies suggest the p53 phospho-
rylation by I3C leading to release p53 and inducing the p21 CDK inhibition and G1
cell cycle arrest [18, 86]. Importantly, treatment with I3C and tamoxifen ablated
expression of the phosphorylated retinoblastoma protein (Rb), an endogenous
140 B. Licznerska and W. Baer-Dubowska
substrate for the G1 CDKs, whereas either agent alone only partially inhibited
endogenous Rb phosphorylation [31]. Several studies showed that I3C and its
derivatives are potent inducers of apoptosis in both ER-positive and ER-negative
breast cancer cells[30–32, 50, 80, 103]. Estrogens, particularly estradiol have been
also implicated as a cofactors in human papillomavirus (HPV)-mediated cervical
cancer, both in animal models and in women using oral contraceptives [72].
Interestingly, it was found that estradiol protects cervical cancer cells treated with
DNA-damaging agents such as UVB, mitomycin-C, and cisplatin, from apoptotic
death. I3C was able to overcome the anti-apoptotic effect of estradiol but only in
higher concentrations. Treatment with I3C resulted in loss of the survival protein
Bcl-2. However, the amount of apoptosis versus survival and the level of Bcl-2
depended on the I3C/estradiol ratio [22]. In HPV16-transgenic mice, which develop
cervical cancer after chronic estradiol exposure, apoptotic cells were detected in
cervical epitheliumonly in mice exposed to estradiol and fed on I3C [20].
Experiments in which cervical cancer HeLa and SiHa cell lines were used,
demonstrated that DIM also exerts antitumor effects on these cells through its
anti-proliferative and pro-apoptotic roles, especially for SiHa cells. The molecular
mechanism for these effects may be related to its regulatory effects on MAPK and
PI3K pathway and apoptosis proteins. Thus, DIM may be considered a preventive
and therapeutic agent against cervical cancer [135]. The ability of inhibiting
spontaneous occurrence of endometrial adenocarcinoma and preneoplastic lesions
by I3C was also demonstrated in female Donryu rats. It was suggested that this
effect was due to induction by I3C estradiol 2-hydroxylation [68]. On the other
hand, promotion of endometrial adenocarcinoma in same strains of rats initiated
with N-ethyl-N’-nitro-N-nitrosoguanidine by I3C was described [133]. DIM was
also found to have a potent cytostatic effect in cultured human Ishikawa endome-
trial cancer cells. This effect was related to the stimulation of TGF-a expression and
activation of TGF-a signal transduction pathway [75].
Another hormone-dependent cancer, which might be affected by I3C, is prostate
cancer, one the most prevalent malignancy in men worldwide and the second
leading cause of male death in Western countries [16]. Androgens play a critical
role in prostate cancer cells growth and survival. Androgens bind to the androgen
receptor (AR), a steroid nuclear receptor, which is translocated into the nucleus and
binds to AREs in the promoter regions of target genes to induce cell proliferation
and apoptosis. Approximately 80–90 % of prostate cancers are dependent on
androgen at initial diagnosis, and endocrine therapy of prostate cancer is directed
toward the reduction of plasma androgens and inhibition of AR [37, 54]. It was
demonstrated that both I3C and DIM are able to downregulate AR signaling [74],
but only DIM was shown to be a strong antagonist of AR and inhibitor of its
translocation to the nucleus [30, 74].
Similarly as in the case of breast cancer cells, I3C and its derivatives also affect
cell cycle progression and induce apoptosis. In this regard, cell cycle arrest at G1
checkpoint in different human prostate carcinoma cell lines by I3C and DIM was
described [2]. In LNCaP prostate cancer cells I3C selectively inhibited the
expression of CDK6 protein and transcripts and stimulated the production of the
6 Indole-3-Carbinol and Its Role in Chronic Diseases 141
p16 CDK inhibitor. In vitro protein kinase assays revealed inhibition by I3C CDK2
enzymatic activity and the relatively minor downregulation of CDK4 enzymatic
activity [134].
In PC-3 cell line induction of G1 cell cycle arrest by I3C due to the upregulation
of p21(WAF1) and p27(Kip1) CDK inhibitors, followed by their association with
cyclin D1 and E and downregulation of CDK6 protein kinase levels and activity
was suggested. In addition, I3C inhibited the hyperphosphorylation of the
retinoblastoma (Rb) protein in PC-3 cells. Induction of apoptosis was also observed
in this cell line when treated with I3C. Thus, it was suggested that I3C inhibits the
growth of PC-3 prostate cancer cells by inducing G1 cell cycle arrest leading to
apoptosis, and regulates the expression of apoptosis-related genes [23]. Further
studies showed that I3C-induced apoptosis is partly mediated by the inhibition of
Akt activation, resulting in the alterations in the downstream regulatory molecules
of Akt activation in PC-3 cells [24]. In the case of DIM an inhibition of a crosstalk
between Akt and NF-jB [12], leading to cell cycle arrest and induction of apoptosis
was also described. DIM significantly decreased cellular histone deacetylase
HDAC2 protein level in androgen sensitive LNCaP and androgen insensitive PC-3
cell lines [10]. In all these studies a formulated DIM (BR-DIM) with higher
bioavailability was used and was able to induce apoptosis and inhibit cell growth,
angiogenesis, and invasion of prostate cancer cells [21].
The potential protective activity of I3C and DIM against prostate cancer was
confirmed by microarray analysis, which showed the modulation of the expression
of many genes related to the control of carcinogenesis and cell survival as effect of
indoles treatment of PC-3 cells [76]. It was also demonstrated by several groups that
I3C and DIM may improve the therapeutic effect of conventional chemotherapy of
prostate cancer [44, 71].
Besides the hormone-dependent cancers, both indoles can affect the develop-
ment of some other cancers. In this regard, it was shown that I3C and DIM induced
apoptosis in colorectal cancer cell lines [13, 67, 84]. Interestingly, an effective
inhibition of Akt and inactivation of mTOR was observed as a result of combined
treatment with I3C and genisteinin HT29 colon cancer cells, leading to induction of
apoptosis and autophagy [93].
Anti-carcinogenic activity of I3C was demonstrated in carcinogen-induced lung
cancer in mice [63, 64]. Anti-proliferative effects of I3C and DIM in human
bronchial epithelial cells (HBEC) and A549 adenocarcinomic human alveolar basal
epithelial cells related to marked reductions in the activation of Akt, extracellular
signal-regulated kinase and NF-jB were also described [65].
Moreover, upregulation of several miRNAs induced by chemical carcinogen was
reversed by I3C in mice and rats lung tumors [46, 59].
The signal transducer and activator of transcription 3 (STAT3) is a latent tran-
scription factor required in proliferation and differentiation. The constitutive acti-
vation of STAT3 in human pancreatic carcinoma specimens but not in normal
tissues was shown. Activation of STAT3 was also found in pancreatic tumor cell
lines and was inhibited by I3C although in relatively high concentration (10 µM)
along with induction of apoptosis [79]. Apoptosis in pancreatic cancer cells was
142 B. Licznerska and W. Baer-Dubowska
Cardiovascular diseases still remain the primary cause of death worldwide. One of
the proposed approaches to reduce the high global incidence is the consumption of
vegetables and fruits containing biologically active components or phytochemical
supplements. Although the most attention was paid to resveratrol, components of
cruciferous vegetables, particularly Brassica oleracea, were also considered a
potential dietary phytochemicals reducing risk of CVDs [97].
In this regard hypocholesterolemic properties of I3C were reported in mice
provided with cholesterol-supplemented diet to which I3C were added. Since
in vitro experiments revealed that I3C and its condensation products effectively
inhibited the enzyme acyl-CoA:cholesterol acyltransferase (ACAT), which is
responsible for the conversion of free cholesterol to the cholesteryl ester, the
hypocholesterolemic effect of I3C in mice was likely mediated by the inhibition of
ACAT [42]. Such mechanism was further confirmed in HepG2 cells. As a result of
treatment with I3C the decreased cholesteryl ester synthesis was associated with
significantly decreased ACAT gene expression and activity [85].
Moreover, antiplatelet and antithrombotic activity of I3C was shown in in vitro
and in vivo studies. I3C significantly inhibited collagen-induced platelet aggrega-
tion in human platelet-rich plasma and suppressed the death of mice with
6 Indole-3-Carbinol and Its Role in Chronic Diseases 143
Moreover, it was shown that in rats bearing the 13762 mammary carcinoma,
addition of I3C to the diet for 6 days prior to antitumor drug ET-743 (trabectidin)
administration almost completely abolished manifestations of hepatotoxicity [41].
These observation further supports the concept that I3C or DIM protecting against
specific cancer may be used in adjuvant therapy to overcome side-effects of con-
ventional therapy.
Specific rodent models like mouse carotid artery injury were developed and used
to assess I3C or DIM protection against cardiovascular diseases, obesity, or dia-
betes. As it was described in previous section, generally the results of these studies
suggest that I3C has a potential benefit in preventing obesity and metabolic dis-
orders, and the action of I3C in vivo may involve multiple mechanisms including
decreased adipogenesis and inflammation, along with activated thermogenesis.
Promising results of the most studies obtained in human cancer cell lines and in
animal models prompted the clinical trials dietary intervention studies to evaluate
the effect of I3C or DIM in risk group of patients or/and volunteers. A major focus
of these trials has been on modulation of hormones metabolism. The urinary
estrogen metabolite ratio of 2-hydroxyesterone to 16a-hydroxyestrone was used in
most of the trials as the surrogate endpoint biomarker.
The validity of this endpoint biomarker was confirmed in the early randomized
clinical trials [14] in which 20 healthy subjects received 400 mg/day of I3C for
3 months. In most of the enrolled subjects I3C increased the 2-hydroxyestrone to
estriol (a precursor of 16a-hydroxyestrone) ratio in sustained manner without
detectible side-effects, although some individuals were resistant to such change. In
another trial women at increased risk for breast cancer were administered with
different doses (range 50–400 mg/day) of I3C for 4 weeks. The results of this study
suggested that the minimum effective dose schedule of 300 mg/day is optimal for
breast cancer prevention, although should be confirmed by long-term breast cancer
prevention trial [128].
In subsequent studies by Bradlow group [91] urine samples were collected from
healthy subjects before and after oral ingestion of 6–7 mg/kg per day for 1 week (7
men) or 2 months (10 women). Analysis of 13 estrogen profiles supported the
hypothesis that I3C induces estrogen 2-hydroxylation resulting in decreased con-
centrations of metabolites known to activate the estrogen receptor and suggested
that I3C may have chemopreventive activity against breast cancer in humans. Later,
phase I trial with women with a high-risk breast cancer were enrolled, subjects
ingested 400 mg I3C daily for 4 weeks followed by a 4 week period of 800 mg I3C
daily [105]. The maximal ratio increase of the urinary 2-hydroxyestrone to
16a-hydroxyestrone was observed with the 400 mg daily dose of I3C, with no
further increase found at 800 mg daily. Beside confirmation of the optimal dose of
146 B. Licznerska and W. Baer-Dubowska
I3C, these studies showed the induction of CYP1A2 which was mirrored by
increase of 2-hydroxyestrone to 16a-hydroxyestrone ratio, and GST.
Cumulative evidence on conversion of I3C to DIM in cell culture, peritoneal and
oral use as well as substantial direct activity seen with DIM led to conclusion that
there is no longer the case for considering I3C to be directly active, and rather DIM
should be considered as a chemopreventive compound of choice [15]. A pilot study
on the effect of BR-DIM on urinary hormone metabolites in postmenopausal
women with a history of early-stage breast cancer showed a significant increase in
levels of 2-hydroxyestrone as result of treatment with only 108 mg DIM/day for
30 days, however, nonsignificant increase (1.46–2.14) of 2-hydroxyestrone to 16a -
hydroxyestrone was noted [33]. In another study cohorts of 3–6 patients
castrate-resistant, non-metastatic prostate cancer received escalating oral doses
twice daily of BR-DIM 75 mg, then 150, 225, and 300 mg. Based on the results of
this trial 225 mg BR-DIM dose twice daily was recommended for phase II trial.
However, modest efficacy of DIM was demonstrated [53].
Cervical intraepithelial neoplasia (CIN) is a precancerous lesion of cervix. When
patients with biopsy proven CIN grade II or III were treated orally with 200, or
400 mg/day of I3C for 12 weeks 50 % of them had complete regression based on
their 12-week biopsy. Moreover, 2-hydroxyestrone to 16a-hydroxyestrone ratio
have changed in a dose-dependent manner [11]. The significant improvement in
confirmed CIN I or II grade was also observed as a result of oral treatment with
2 mg/kg/day of DIM for 12 weeks. Moreover, at median follow-up of 6 months
there was no statistically significant difference in any of the measured outcome
between the DIM and placebo group [40].
Since the incidence of thyroid cancer is 4–5 times higher in women than in men,
estrogens were suggested to contribute the pathogenesis of thyroid proliferative
disease (TPD). In limited (7 patients) phase I clinical trial patients with TPD were
administered with 300 mg of DIM per day for 14 days. DIM was detectable in
thyroid tissue, and the ratio of 2-hydroxyestrone to 16a-hydroxyestrone was
increased. These results suggested that DIM can manifest the anti-estrogenic
activity in situ to modulate TPD [104].
Although major focus of cancer prevention clinical trials of I3C or DIM has been
concentrated on chemoprevention of hormone-dependent cancers, there were also
clinical trials performed in order to evaluate indoles effect on pulmonary cancers. In
this regard in phase I clinical trial patients with recurrent respiratory papillomatosis
(RRP) were treated orally with I3C and had minimum follow-up of 8 months.
Thirty-three percent of the study patients had a cession of their papilloma growth
and had not required surgery since the start of the study [107]. Subsequent
long-term clinical trial performed by the same research group confirmed the pre-
liminary observation indicating that I3C may be a treatment option for RRP [108].
The case of successful use of intralesional and intravenous cidofovir in association
with I3C in 8-year-old girl with pulmonary papillomatosis was also reported [38].
As it was mentioned in the previous sections of this chapter, recently a large
amount of evidence has demonstrated that epigenetic alterations, such as DNA
6 Indole-3-Carbinol and Its Role in Chronic Diseases 147
supplements should be restricted until potential risks and benefits are better char-
acterized. Taking into consideration higher activity of DIM, particularly in BR
form, in comparison with I3C in term of potency and time required to obtain
the effect, this I3C dimer might be a better alternative as chemopreventive sup-
plement. Important aspect of possible clinical application of both indoles is their
drug and radio-sensitization. Emerging new technologies allowing deeper inside in
the mechanism of these glucosinolate derivatives activity should help to better
explore this aspect.
References
56. Horn TL, Reichert MA, Bliss RL (2002) Modulations of P450 mRNA in liver and mammary
gland and P450 activities and metabolism of estrogen in liver by treatment of rats with
indole-3-carbinol. Biochem Pharmacol 64:393–404
57. Hwang JW, Jung JW, Lee YS et al (2008) Indole-3-carbinol prevents H(2)O(2)-induced
inhibition of gap junctional intercellular communication by inactivation of PKB/Akt. J Vet
Med Sci 70:1057–1063
58. International Agency for Research on Cancer (1999) Monographs on the evolution of
carcinogenic risks to humans: hormonal contraception and postmenopausal hormone therapy,
vol 72. IARC, Lyon, France
59. Izzotti A, Calin GA, Steele VE et al (2010) Chemoprevention of cigarette smoke-induced
alterations of microRNA expression in rat lungs. Cancer Prev Res 3:62–72
60. Jayakumar P, Pugalendi KV, Sankaran M (2014) Attenuation of hyperglycemia-mediated
oxidative stress by indole-3-carbinol and its metabolite 3, 3’- diindolylmethane in C57BL/6 J
mice. J Physiol Biochem 70:525–534
61. Jin L, Qi M, Chen DZ et al (1999) Indole-3-carbinol prevents cervical cancer in human
papilloma virus type 16 (HPV16) transgenic mice. Cancer Res 59:3991–3997
62. Jin Y (2011) 3,3’-Diindolylmethane inhibits breast cancer cell growth via miR-21-mediated
Cdc25A degradation. Mol Cell Biochem 358:345–354
63. Kassie F, Anderson LB, Scherber R et al (2007) Indole-3-carbinol inhibits 4-
(methylnitrosamino)-1-(3-pyridyl)-1-butanone plus benzo(a)pyrene-induced lung
tumorigenesis in A/J mice and modulates carcinogen-induced alterations in protein levels.
Cancer Res 67:6502–6511
64. Kassie F, Kalscheuer S, Matise I et al (2010) Inhibition of vinyl carbamate-induced
pulmonary adenocarcinoma by indole-3-carbinol and myo-inositol in A/J mice.
Carcinogenesis 31:239–245
65. Kassie F, Melkamu T, Endalew A et al (2010) Inhibition of lungcarcinogenesis and critical
cancer-related signaling pathways by N-acetyl-S-(N-2-phenethylthiocarbamoyl)-l-cysteine,
indole-3-carbinol and myo-inositol, alone and in combination. Carcinogenesis 31:1634–1641
66. Kim DJ, Han BS, Ahn B et al (1997) Enhancement by indole-3-carbinol of liver and thyroid
gland neoplastic development in a rat medium-term multiorgan carcinogenesis model.
Carcinogenesis 18:377–381
67. Kim EJ, Park Sy, Shin et al (2007) Activation of caspase-8 contributes to 3,3’-
Diindolylmethane-induced apaptosis in colon cancer cells. J Nutr 137:31–36
68. Kojima T, Tanaka T, Mori H (1994) Chemoprevention of spontaneous endometrial cancer in
female Donryu rats by dietary indole-3-carbinol. Cancer Res 54:1446–1449
69. Kong D, Heath E, Chen W et al (2012) Loss of let-7 up-regulates EZH2 in prostate cancer
consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM.
PLoS ONE 7:e33729
70. Kong D, Heath E, Chen W et al (2012) Epigenetic silencing of miR-34a in human prostate
cancer cells and tumor tissue specimens can be reversed by BR-DIM treatment. Am J Transl
Res 4:14–23
71. Kumi-Diaka J, Merchant K, Haces A et al (2010) Genistein-selenium combination induces
growth arrest in prostate cancer cells. J Med Food 13:842–850
72. Kumar MM, Davuluri S, Poojar S et al (2015) Role of estrogen receptor alpha in human
cervical cancer-associated fibroblasts: a transcriptomic study. Tumour Biol Oct 24 [Epub
ahead of print]
73. Lawrence T (2009) The nuclear factor NF-jB pathway in Inflammation. Cold Spring Harb
Perspect Biol 1:a001651. doi:10.1101/cshperspect.a001651
74. Le HT, Schaldach CM, Firestone GL et al (2003) Plant-derived 3,3’-Diindolylmethane is a
strong androgen antagonist in human prostate cancer cells. J Biol Chem 278:21136–21145
75. Leong H, Riby JE, Firestone GL et al (2004) Potent ligand-independent estrogen receptor
activation by 3,3′-diindolylmethane is mediated by cross talk between the protein kinase A
and mitogen-activated protein kinase signaling pathways. Mol Endocrinol 18:291–302
152 B. Licznerska and W. Baer-Dubowska
76. Li Y, Li X, Sarkar FH (2003) Gene expression profiles of I3C- and DIM-treated PC3 human
prostate cancer cells determined by cDNA microarray analysis. J Nutr 133:1011–1019
77. Li Y, Wang Z, Kong D et al (2007) Regulation of FOXO3a/beta-catenin/GSK-3beta
signaling by 3,3’-diindolylmethane contributes to inhibition of cell proliferation and
induction of apoptosis in prostate cancer cells. J Biol Chem 282:21542–21550
78. Li Y, VandenBoomII TG, Wang Z et al (2010) miRNA146a suppresses invasion of
pancreatic cancer cells. Cancer Res 70:1486–1495
79. Lian JP, Word B, Taylor S et al (2004) Modulation of the constitutive activated STAT3
transcription factor in pancreatic cancer prevention: effects of indole-3-carbinol (I3C) and
genistein. Anticancer Res 24:133–137
80. Licznerska BE, Szaefer H, Murias M et al (2013) Modulation of CYP19 expression by
cabbage juices and their active components: indole-3-carbinol and 3,3’-diindolylmethane in
human breast epithelial cell lines. Eur J Nutr 52:1483–1492
81. Lo R, Matthews J (2013) The aryl hydrocarbon receptor and estrogen receptoralpha
differentially modulate nuclear factor erythroid-2-related factor2 transactivation in MCF-7
breast cancer cells. Toxicol Appl Pharmacol 270:139–148
82. Lu Q, Nakmura J, Savinov A et al (1996) Expression of aromatase protein and messenger
ribonucleic acid in tumor epithelial cells and evidence of functional significance of locally
produced estrogen in human breast cancer. Endocrinology 137:3061–3068
83. Luo J, Manning BD, Cantley LC (2003) Targeting the PI3 K-Akt pathway in human cancer:
rationale andpromise. Cancer Cell 4:257–262
84. Lynn A, Collins A, Fuller Z et al (2006) Cruciferous vegetables and colorectal cancer. Proc
Nutr Soc 65:135–144
85. Maiyoh GK, Kuh JE, Casaschi A et al (2007) Cruciferous indole-3-carbinol inhibits
apolipoprotein B secretion in HepG2 cells. J Nutr 137:2185–2189
86. Marconett CN, Singhal AK, Sundar SN et al (2012) Indole-3-carbinol disrupts estrogen
receptor-alpha dependent expression of insulin-like growth factor-1 receptor and insulin
receptor substrate-1 and proliferation of human breast cancer cells. Mol Cell Endocrinol
363:74–84
87. McGuire KP, Ngoubilly N, Neavyn M et al (2006) 3,3′-diindolylmethane and paclitaxel act
synergistically to promote apoptosis in HER2/Neu human breast cancer cells. J Surg Res
132:208–213
88. Melkamu T, Zhang X, Tan J et al (2010) Alteration of microRNA expression in vinyl
carbamate-induced mouse lung tumors and modulation by the chemopreventive agent
indole-3-carbinol. Carcinogenesis 31:252–258
89. Meng Q, Qi M, Chen DZ et al (2000) Suppression of breast cancer invasion and migration by
indole-3-carbinol: associated with up-regulation of BRCA1 and E-cadherin/catenin
complexes. J Mol Med 78:155–165
90. Mesnil M, Crespin S, Avanzo JL et al (2005) Defective gap junctional intercellular
communication in the carcinogenic process. Biochim Biophys Acta 1719:125–145
91. Michnovicz JJ, Adlercreutz H, Bradlow HL (1997) Changes in levels of urinary estrogen
metabolites after oral indole-3-carbinol treatment in humans. J Natl Cancer Inst 89:718–723
92. Mulvey L, Chandrasekaran A, Liu K et al (2007) Interplay of genes regulated by estrogen
and diindolylmethane in breast cancer cell lines. Mol Med 13:69–78
93. Nakamura Y, Yogosawa S, Izutani Y et al (2009) A combination of indol-3-carbinol and
genistein synergistically induces apoptosis in human colon cancer HT-29 cells by inhibiting
Aktphosphorylation and progression of autophagy. Mol Cancer 8:100. doi:10.1186/1476-
4598-8-100
94. Nachshon-Kedmi M, Yannai S, Haj A et al (2003) Indole-3-carbinol and 3,3’-
diindolylmethane induce apoptosis in human prostate cancer cells. Food Chem Toxicol
41:745–752
95. Oganesian A, Hendricks JD, Williams DE (1997) Long term dietary indole-3-carbinol
inhibits diethylnitrosamine-initiated hepatocarcinogenesis in the infant mouse model. Cancer
Lett 118:87–94
6 Indole-3-Carbinol and Its Role in Chronic Diseases 153
118. van Poppel G, Verhoeven DT, Verhagen H et al (1999) Brassica vegetables and cancer
prevention. Epidemiology and mechanisms. Adv Exp Med Biol 472:159–168
119. Vahid F, Zand H, Nosrat-Mirshekarlou E et al (2015) The role dietary of
bioactivecompounds on the regulation of histone acetylases and deacetylases: a review.
Gene 562:8–15
120. Vang O (2006) Chemopreventive potential of compounds in Cruciferous vegetables. In:
Baer-Dubowska W, Bartoszek A, Malejka-Giganti D (eds) Carcinogenic and
anticarcinogenic food components. CRC Taylor & Francis, Boca Raton, pp 303–328
121. Verhagen H, Poulsen HE, Loft S et al (1995) Reduction of oxidative DNA-damage in
humans by brussels sprouts. Carcinogenesis 16:969–970
122. Wattenberg LW, Loub WD (1978) Inhibition of polycyclic aromatic hydrocarbon-induced
neoplasia by naturally occurring indoles. Cancer Res 38:1410–1413
123. Wattenberg LW, Loub WD, Lam LK, Speier JL (1976) Dietary constituents altering the
responses to chemical carcinogens. Fed Proc 35:1327–1331
124. Wattenberg LW, Hanley AB, Barany G et al (1985) Inhibition of carcinogenesis by some
minor dietary constituents. Princess Takamatsu Symp 16:193–203
125. WHO Report Part II 2015
126. Wilson CA, Ramos L, Villaseñor MR et al (1999) Localization of human BRCA1 and its loss
in high-grade, non-inherited breast carcinomas. Nat Genet 21:236–240
127. Witter DC, Le Bas J (2008) Cancer as a chronic disease. Oncology 53:1–3
128. Wong GY, Bradlow L, Sepkovic D et al (1997) Dose-ranging study of indole-3-carbinol for
breast cancer prevention. J Cell Biochem Suppl 28–29:111–116
129. Wong CP, Hsu A, Buchanan A et al (2014) Effects of sulforaphane and 3,3′-
diindolylmethane on genome-wide promoter methylation in normalprostate epithelial cells
and prostate cancer cells. PLoS ONE 9:e86787. doi:10.1371/journal.pone.0086787
130. Wu TY, Khor TO, Su ZY et al (2013) Epigenetic modifications of Nrf2 by 3,3′-
diindolylmethanein vitro in TRAMP C1 cell line and in vivo TRAMP prostate tumors.
AAPS J 15:864–874
131. Xu M, Orner GA, Bailey GS et al (2001) Post-initiation effects of chlorophyllin and
indole-3-carbinol in rats given 1,2-dimethylhydrazine or 2-amino-3- methylimidazo[4, 5-f]
quinoline. Carcinogenesis 22:309–314
132. Xu H, Barnes GT, Yang Q et al (2003) Chronic inflammation in fat plays a crucial role in the
development of obesity-related insulin resistance. J Clin Invest 112:1821–1830
133. Yoshida M, Katashima S, Ando J et al (2004) Dietary indole-3-carbinol promotes
endometrial adenocarcinoma development in rats initiated with N-ethyl-N’-nitro-N-
nitrosoguanidine, with induction of cytochrome P450 s in the liver and consequent
modulation of estrogen metabolism. Carcinogenesis 25:2257–2264
134. Zhang J, Hsu BAJC, Kinseth BAMA et al (2003) Indole-3-carbinol induces a G1 cell cycle
arrest and inhibits prostate-specificantigen production in human LNCaP prostate carcinoma
cells. Cancer 98:2511–2520
135. Zhu J, Li Y, Guan C et al (2012) Anti-proliferative and pro-apoptotic effects of 3, 3’-
diindolylmethane in human cervical cancer cells. Oncol Rep 28:1063–1068
Chapter 7
Sanguinarine and Its Role in Chronic
Diseases
Abstract The use of natural products derived from plants as medicines precedes
even the recorded human history. In the past few years there were renewed interests
in developing natural compounds and understanding their target specificity for drug
development for many devastating human diseases. This has been possible due to
remarkable advancements in the development of sensitive chemistry and biology
tools. Sanguinarine is a benzophenanthridine alkaloid derived from rhizomes of the
plant species Sanguinaria canadensis. The alkaloid can exist in the cationic imi-
nium and neutral alkanolamine forms. Sanguinarine is an excellent DNA and RNA
intercalator where only the iminium ion binds. Both forms of the alkaloid, however,
shows binding to functional proteins like serum albumins, lysozyme and hemo-
globin. The molecule is endowed with remarkable biological activities and large
number of studies on its various activities has been published potentiating its
development as a therapeutic agent particularly for chronic human diseases like
cancer, asthma, etc. In this article, we review the properties of this natural alkaloid,
and its diverse medicinal applications in relation to how it modulates cell death
signaling pathways and induce apoptosis through different ways, its utility as a
therapeutic agent for chronic diseases and its biological effects in animal and human
models. These data may be useful to understand the therapeutic potential of this
important and highly abundant alkaloid that may aid in the development of
sanguinarine-based therapeutic agents with high efficacy and specificity.
Keywords Benzophenanthridine Sanguinarine Nucleic acid binding property
Anticancer Antimicrobial
7.1 Introduction
Sanguinarine[13-methyl[1,3]benzodioxolo[5,6-c]-1,3-dioxolo[4,5-i]phenanthridinium]
(Fig. 7.1) is the well-known member of the relatively small group of quaternary benzo
[c]phenanthridine [QBA] alkaloids. Sanguinaria canadensis, also known as bloodroot,
is a perennial herbaceous flowering plant of the papaveriaceae family. Bloodroot pro-
duces primarily the toxic alkaloid sanguinarine that is largely stored in the rhizome of
the plant. From a medicinal perspective, QBAs in general and sanguinarine in particular,
have many important properties. They display antimicrobial, antifungal and
anti-inflammatory effects in addition to their putative anticancer activity that is widely
studied. The molecular action of sanguinarine in expressing its biological activity has
been a subject of intense debate; it has been shown to interact with many targets, like
nucleic acids, proteins and microtubules, and modify the activities of a wide variety of
enzymes. This review summarizes the current state of knowledge on the properties
of sanguinarine that are important for their potential use in chronic diseases.
Sanguinarine occurs either as chloride or sulfate crystalline salts and both are
orange-red colored. It is sparingly soluble in aqueous conditions but highly soluble
in many organic solvents. Some of the physical properties of sanguinarine are
summarized in Table 7.1.
One of the remarkable properties of sanguinarine and other QBAs is their ability
to exhibit a pH dependent structural equilibrium between the quaternary iminium
form and the 6-hydroxydihydro derivative or the alkanolamine form (Scheme 7.1).
The reversible, pH dependent equilibrium between the charged iminium (SGI) and
the neutral alkanolamine (SGA) forms in aqueous solution may be represented as
follows [76]
SG þ þ OH $ SGOH
36, 39, 42]. These two forms have characteristic absorbance and fluorescence
spectra which are presented in Fig. 7.2a, b. Thus, in aqueous solution at physio-
logical pH both forms of the alkaloid are present. The SGI is unsaturated and
planar; the SGA has a tilted structure and is essentially nonplanar. The aqueous
solubility of the SGI is much higher than that of the SGA. The latter (SGA) form
can penetrate the cell membrane increasing its cellular availability. Inside the cell it
may be converted partially to the SGI form influenced by pH and other factors. Due
to the conjugated aromatic nature of its chromophore sanguinarine has strong
spectral bands in the UV–vis region and also exhibits intense fluorescence
(Fig. 7.2b). The molecule is, therefore, sensitive to light and can cause adverse
phototoxic reactions in the presence of light. The SGA form is known to undergo
photo oxidation at the C6 carbon atom to oxysanguinarine in alkaline solutions
(Scheme 7.2) [75]. The photo oxidation of sanguinarine was found to be acceler-
ated in the presence of Rose Bengal suggesting the role of singlet molecular oxygen
in the reaction mechanism [28].
158 P. Basu and G.S. Kumar
The toxicity of sanguinarine has been suggested to be due to its DNA interca-
lation, inhibition of ion pumps and several thiol dependent proteins, and interaction
with cyto skeletal components [24, 86]. Sanguinarine can kill animal cells through a
variety of mechanisms. Epidemic dropsy is a form of edema of extremities that
results from ingesting argemone oil that contains sanguinarine [73]. Application of
sanguinarine to skin may result in tissue damage because of escharotic effect.
Phototoxic effect of sanguinarine against mosquito larvae has also been reported
[67]. Production of singlet oxygen by sanguinarine is known [6, 54]. Phototoxic
action of sanguinarine has also been suggested to be due to the production of H2O2
[6, 78]. Reactive oxygen species (ROS) such as superoxide anions (O2−) or
7 Sanguinarine and Its Role in Chronic Diseases 159
Scheme 7.2 Generation of oxysanguinarine from SGA form in the presence of oxygen by
photochemical process. Reprinted from Suresh Kumar et al. 1997 [75] with permission. Copyright
Elsevier Science S. A
Fig. 7.3 Sanguinarine molecule intercalated at the interface of two “two molecules” DNA units.
DNA chains color scheme: A = orange, B = yellow, C = green, D = blue. Reprinted from [25]
with permission. Copyright The Royal Society of Chemistry 2011
Sanguinarine has been reported to induce cell cycle arrest and apoptosis in distinct
cancer cells [3, 4, 20, 32, 45, 72, 85]. It modulates multiple signaling pathways
causing inhibition of the initiation of cancer, inducing cell cycle arrest, apoptosis,
and inhibiting metastasis and angiogenesis in a variety of cancer cells [9, 82]
(Fig. 7.4).
One of the first systematic study showing the anticancer effect of sanguinarine
was that of Ahmad et al. [4] who showed that sanguinarine treatment resulted in an
apoptotic death of human epidermoid carcinoma A431 carcinoma cells and the loss
of viability that occurred at much less losses in comparison to normal human
epidermal keratinocytes (NHEKs). The DNA cell cycle analysis revealed that
sanguinarine treatment did not significantly affect the distribution of cells among
the different phases of the cell cycle in A431 cells. It was suggested that the
involvement of the NFĸB pathway may be a mechanism of sanguinarine-mediated
apoptosis [4]. A study by Ding et al. [20] reported that sanguinarine-induced
concentration dependent apoptosis with caspase-3 activation and BCD/oncosis
without caspase-3 activation suggesting two cell death mechanisms. This suggests
7 Sanguinarine and Its Role in Chronic Diseases 161
Fig. 7.4 Schematic diagram showing the multiple cell signaling pathway of action of
sanguinarine in modulating anticancer activity
and androgen-unresponsive (DU145) human prostate cancer cells and that this
effect was mediated through deregulation of cell cycle and induction of apoptosis.
The involvement of cyclin kinase inhibitors (cki) and cyclin-dependent kinases
(cdk), i.e., the cki–cyclin–cdk machinery, during cell cycle arrest and apoptosis of
prostate cancer cells by sanguinarine has been suggested [3].
Sanguinarine was found not increase Bax levels in K1735-M2 melanoma cells.
A similar result was obtained in human CEM T-leukemia cells [43]. The interesting
result was that the absence of Bax over-expression occurred with an increase of p53
protein. Matkar et al. [59] demonstrated that sanguinarine-induced death in human
colon cancer cells was p53-independent, although DNA damage was detected in
K1735-M2 cells. The results suggested that mitochondrial depolarization induced
by sanguinarine is associated with nuclear DNA damage, although it may not
exclusively result from pathways activated by the latter event [72].
In vascular smooth muscle cells significant growth inhibition was induced by
sanguinarine as a result of G1-phase cell cycle arrest mediated by induction of
p27KIP1 expression, and resulted in a down-regulation of the expression of cyclins
and cdks. Moreover, sanguinarine-induced inhibition of cell growth appeared to be
linked to activation of Ras/ERK through p27KIP1-mediated G1-phase cell cycle
arrest. Overall, the unexpected effects of sanguinarine treatment in VSMC provided
a theoretical basis for clinical use of therapeutic agents in the treatment of
atherosclerosis [49].
Some researches ascribe the pro-apoptotic properties of sanguinarine to pro-
duction of ROS [29, 30, 43] while others have demonstrated that the effects of
sanguinarine are not accompanied by ROS production [19, 60, 81]. Kim et al. [46]
reported that the mechanism of sanguinarine-induced apoptosis in human breast
cancer MDA-231 cells was due to the generation of ROS. Its ability to directly
interact with glutathione (GSH) was thought to lead to depletion of cellular GSH
and induction of ROS generation [12, 19, 38, 46]. It was found that the quenching
of ROS generation by N-acetyl-L-cysteine (NAC), a scavenger of ROS, reversed
sanguinarine-induced apoptosis effects. It was also found that sanguinarine-induced
rat hepatic stellate T6 cells (HSC-T6 cells) apoptosis was correlated with the
generation of increased ROS, leading to decrease in the mitochondrial membrane
potential (MMP) and the down-regulation of anti-apoptotic protein Bcl-2 [88].
Sanguinarine is a selective inhibitor of mitogen-activated protein kinase phos-
phatase 1 (MKP-1), which is over expressed in many tumor cells [79]. The dis-
ruption of microtubule assembly dynamics [51], the nucleocytoplasmic trafficking
of cyclin D1 and Topisomerase II [32] and the induction of DNA damage [18]
allosteric activator of AMP-activated protein kinase [14] also contributed, at least in
part to the anticancer effects of sanguinarine [52].
Sanguinarine induces a rapid caspase-dependent cell death in human melanoma
cells; partially involving endoplasmic reticulum and mitochondria mediated
responses. This cell death is dependent on the generation of reactive oxygen spe-
cies, and is not prevented by Bcl-XL over expression. The fact that sanguinarine
induces a very rapid cell death with apoptotic features in melanoma cells, together
with the lack of inhibition by over expressing Bcl-XL, highlight the potent
7 Sanguinarine and Its Role in Chronic Diseases 163
anti-melanoma activity of this isoquinoline alkaloid and suggest its potential in the
treatment of skin cancer [9]. Recently Singh et al. [74] identified a number of
differentially expressed proteins which are important in the signaling pathways
modulated by sanguinarine in its action against pancreatic cancer.
Chronic diseases, like heart disease, COPD, stroke, cancer, respiratory diseases like
asthma and metabolic diseases like diabetes are the leading cause of human mor-
tality. Many natural products in general and sanguinarine in particular possess
potent activities that have been useful in the treatment of these chronic ailments,
and these have also been documented in many studies. Anti proliferative activities
have been demonstrated in cells derived from various human carcinomas as
reviewed in the previous section. Various mechanisms by which sanguinarine
induce apoptosis in cancer cells through various pathways have been described. The
therapeutic potential of sanguinarine in cardiovascular disease related to platelet
aggregation has been reported by Jeng et al. [41, 53]. Sanguinarine has promising
antihypertensive and potent antiplatelet effects; it interferes with renin-angiotensin
system and possibly other blood pressure regulating pathways, and also induces
calcium mobilization, thromboxane and cAMP production (Scheme 7.3). Many
studies on in vitro models are reported to this effect and a recent review summarizes
the antihypertensive activity and cardiovascular properties, including hypotensive,
antiplatelet, and positive inotropic effects of sanguinarine [53]. Computational
bioinformatics analysis has identified sanguinarine as a potential candidate drugs
for the treatment of type 2 diabetes [83] but clinical studies are not yet available on
this aspect.
Scheme 7.3 Schematic diagram showing the role of sanguinarine in different chronic diseases
164 P. Basu and G.S. Kumar
HT22 cells from apoptotic cell death induced by glutamate. Sanguinarine signifi-
cantly attenuated the loss of mitochondrial function and membrane integrity asso-
ciated with glutamate-induced neurotoxicity. Sanguinarine protected against
glutamate-induced neurotoxicity through inhibition of HT22 cell apoptosis. JC-1
staining, a well-established measure of mitochondrial damage, which decreased
after treatment with sanguinarine in glutamate-challenged HT22 cells. In addition,
sanguinarine diminished the intracellular accumulation of ROS and Ca2+.
Non-cytotoxic concentration of sanguinarine led to a marked protection against
glutamate-induced oxidative toxicity in mice through activation of the Nrf2-HO-1
pathway. The antioxidant effect of HO-1 was mainly due to reduction in ROS
production and recovery of mitochondrial decay that promoted a decrease in
glutamate-trigged apoptosis in HT22 cells [65].
An antidepressant-like effect of sanguinarine in rats was reported through
inhibition of mitogen-activated protein kinase phosphatase-1 in rates that enable the
use of sanguinarine as anti depressant drug [13]. Radiation protective efficacy of
sanguinarine in mice models have also been reported [87].
Recently, sanguinarine treatment was shown to result in reduction of cell
migration, a dose-dependent inhibition of cell viability and the induction of cell
death by apoptosis in both human (MDA-MB-231 cells) and mouse (A17 cells)
in vitro models of basal-like breast cancer (BLBC). Oral administration of san-
guinarine reduced the development and growth of A17 transplantable tumors in
FVB syngeneic mice. The suppression was correlated with a concurrent up regu-
lation of p27 and down-regulation of cyclin D1 and with the inhibition of STAT3
activation.
The adverse effect, viz., the epidemic dropsy syndrome, is a well-known toxic
effect on human health linked to development of glaucoma due to consumption of
edible oil adulterated with argemone oil that principally contained sanguinarine.
This involves initiation of oxidative stress (ROS formation leading to lipid per-
oxidation, depletion of GSH and decrease of total antioxidant capacity) and death of
red cells via formation of methemoglobin [7, 8, 17]. The detoxification pathway of
sanguinarine in animals and in man may be by conversion to dihydrosanguinarine
which has been reported in rats [22]. On the other hand, in several European Union
nations QBAs and principally sanguinarine are used as feed additives for Pigs.
Sangrovit®, a phytogenic feed additive derived from the rhizomes of Sanguinaria
canadensis, contains high amounts of sanguinarine. This has been shown to
increase feed intake and feed conversion in growing pigs, which results in improved
growth performance and stimulates anti-inflammatory activity [44]. A study to
detect sanguinarine-induced DNA damage has indicated the absence of any DNA
adducts in pigs fed with sanguinarine. The observation that the animals remained
healthy contradicted the cause of epidemic dropsy syndrome to sanguinarine [47].
Also broilers treated with drinking water containing sanguinarine showed reduced
Salmonella Enteritidis count [68].
166 P. Basu and G.S. Kumar
Not many studies have been performed in humans on the effect of sanguinarine as
such, but herbal preparations containing sanguinarine have been used in folk
medicines in small doses for the treatment of many disorders like bronchial prob-
lems, asthma, cough and cold remedies, severe throat infections and heart diseases
[26]. Benefits have also been reported for leprosy and tuberculosis treatment,
antimicrobial treatment for the gastrointestinal system, etc. Sanguinarine, as
described in the above section, is traditionally used in tooth pastes and antiseptic
mouth rinses due to its ability to suppress dental plaque gingival inflammation and
periodontal disease through antimicrobial action and ability to inhibit bacterial
adherences [27, 48, 66]. The potential use of sanguinarine in clinical treatment of
allergic asthma has also been suggested from an analysis of gene expression profile
of asthma patients [84]. Sanguinarine is reported to have hERG potassium channel
blocking effect. Therefore, in some situations where cardiac repolarization reserve
is weak, it may increase the risk of potentially fatal ventricular arrhythmias in
canines [63]. The cardiovascular properties of sanguinarine including hypotensive,
antiplatelet and positive inotropic effects have also been described [53]. In a number
of human tumor cells sanguinarine treatment resulted in boosting of intra cellular
ROS, elevation of mitogen-activated protein kinase p38, triggering caspase 3/7
activation, inhibition of mitogen-activated protein kinase phosphatase 1 (MKP-1),
depletion of glutathione and a host of other activities occurred.
There are concerns that the use of sanguinarine may cause leukoplakia in the mouth
and oral dysplastic lesions. The use of sanguinarine and blood root for periodontal
disease abacterially elicited inflammation of the gingival and periodontal tissue is
also not based on adequate scientific support. There are unconfirmed reports that
suggested that sanguinarine may cause sedation, faintness, vertigo, and possibly
impair decision-making and increase response time. Bloodroot was reported to be
used to stimulate menstruation and hence it was not advised to be used during
pregnancy [77].
7.8 Conclusions
inhibit bacterial adherence due to its antimicrobic properties potentiates its clinical
use in periodontitis treatment and mouth washes. Sanguinarine is used as a dietary
supplementation in animal feeds to reduce amino acid degradation, increase feed
intake, and promote growth. Sanguinarine is a putative anticancer agent that is
reported to arrest the cell cycle and induce apoptosis through various mechanisms
that may include binding to DNA and tubilin and inhibiting many enzymes.
Specifically, results from several studies reviewed above have indicated that san-
guinarine inhibits the proliferation of cancer cells of different origins and apoptosis
induction in different malignant cell lines takes place through the activation of cell
surface receptors, intrinsic cytochrome c release from mitochondria pathways, etc.
In spite of a number of studies the precise mechanisms and the diverse pathways by
which the anticancer effect of this compound is manifested are still not fully
understood. The information reviewed and summarized in this chapter reveals that
sanguinarine is a potential cancer drug candidate and detailed studies including
further vivo experiments are indispensable to elucidate whether administration of
sanguinarine as therapeutics would be safe and effective for the treatment of human
diseases.
Acknowledgments GSK acknowledges the Council of Scientific and Industrial Research (CSIR),
Govt. of India for funding the research activities through network projects NWP0036 and
GenCODE (BSC0123). GSK sincerely thanks Dr. M. Maiti, Ex. Scientist of CSIR-Indian Institute
of Chemical Biology, who initiated and introduced him to the research on sanguinarine at the
Biophysical Chemistry Laboratory, and also all the graduate students who have contributed to the
understanding of the chemistry and biochemistry of the alkaloid. PB is a NET-Senior Research
Fellow of the University Grants Commission, Govt. of India.
References
8. Babu CK, Khanna SK, Das M (2007) Adulteration of mustard cooking oil with argemone oil:
Do Indian food regulatory policies and antioxidant therapy both need revisitation? Antioxid
Redox Signal 9:515–525
9. Burgeiro A, Bento AC, Gajate C, Oliveira PJ, Mollinedo F (2013) Rapid human melanoma cell
death induced by sanguinarine through oxidative stress. Eur J Pharmacol 705(1–3):109–118
10. Chan WH (2011) Embryonic toxicity of sanguinarine through apoptotic processes in mouse
blastocysts. Toxicol Lett 205(3):285–292
11. Chan WH (2015) Hazardous effects of sanguinarine on maturation of mouse oocytes,
fertilization, and fetal development through apoptotic processes. Environ Toxicol 30(8):946–955
12. Chang MC, Chan CP, Wang YJ, Lee PH, Chen LI, Tsai YL, Lin BR, Wang YL, Jeng JH
(2007) Induction of necrosis and apoptosis to KB cancer cells by sanguinarine is associated
with reactive oxygen species production and mitochondrial membrane depolarization. Toxicol
Appl Pharmacol 218:143–151
13. Chen Y, Wang H, Zhang R, Wang H, Peng Z, Sun R, Tan Q (2012) Microinjection of
sanguinarine into the ventrolateral orbital cortex inhibits Mkp-1 and exerts an
antidepressant-like effect in rats. Neurosci Lett 506(2):327–331
14. Choi J, He N, Sung MK, Yang Y, Yoon S (2011) Sanguinarine is an allosteric activator of
AMP-activated protein kinase. Biochem Biophys Res Commun 413(2):259–263
15. Chowdhury SR, Islam MM, Suresh Kumar G (2010) Binding of the anticancer alkaloid
sanguinarine to double stranded RNAs: insights into the structural and energetics aspects. Mol
BioSyst 6(7):1265–1276
16. Choy CS, Cheah KP, Chiou HY, Li JS, Liu YH, Yong SF, Chiu WT, Liao JW, Hu CM (2008)
Induction of hepatotoxicity by sanguinarine is associated with oxidation of protein thiols and
disturbance of mitochondrial respiration. J Appl Toxicol 28:945–956
17. Das M, Babu K, Reddy NP, Srivastava LM (2005) Oxidative damage of plasma proteins and
lipids in epidemic dropsy patients: alterations in antioxidant status. Biochim Biophys Acta
1722:209–217
18. De Stefano I, Raspaglio G, Zannoni GF, Travaglia D, Prisco MG, Mosca M, Ferlini C,
Scambia G, Gallo D (2009) Antiproliferative and antiangiogenic effects of the
benzophenanthridine alkaloid sanguinarine in melanoma. Biochem Pharmacol 78(11):1374–
1381
19. Debiton E, Madelmont JC, Legault J, Barthomeuf C (2003) Sanguinarine-induced apoptosis is
associated with an early and severe cellular glutathione depletion. Cancer Chemother
Pharmacol 51:474–482
20. Ding Z, Tang SC, Weerasinghe P, Yang X, Pater A, Liepins A (2002) The
alkaloid sanguinarine is effective against multidrug resistance in human cervical cells via
bimodal cell death. Biochem Pharmacol 63(8):1415–1421
21. Dong XZ, Zhang M, Wang K, Liu P, Guo DH, Zheng XL, Ge XY (2013) Sanguinarine
inhibits vascular endothelial growth factor release by generation of reactive oxygen species in
MCF-7 human mammary adenocarcinoma cells. BioMed Res Int 517698
22. Dvorák Z, Simánek V (2007) Metabolism of sanguinarine: the facts and the myths. Curr Drug
Metab 8(2):173–176
23. Eversole LR, Eversole GM, Kopcik J (2000) Sanguinaria-associated oral leukoplakia.
Comparison with other benign and dysplastic leukoplakic lesions. Oral Surg Oral Med Oral
Pathol Oral Radiol Endod 89:455–464
24. Faddeeva MD, Beliaeva TN (1997) Sanguinarine and ellipticine cytotoxic alkaloids isolated
from well-known antitumor plants: intracellular targets of their action. Tsitologiia 39:181–208
25. Ferraroni M, Bazzicalupi C, Bilia AR, Gratteri P (2011) X-Ray diffraction analyses of the
natural isoquinoline alkaloids berberine and sanguinarine complexed with double helix DNA d
(CGTACG). Chem Commun 47(17):4917–4919
26. Frankos VH, Brusick DJ, Johnson EM, Maibach HI, Munro I, Squire RA, Weil CS (1990)
Safety of sauguinaria extract as used in commercial tooth- paste and oral rinse products. J Can
Dent Assoc 56:41–47
7 Sanguinarine and Its Role in Chronic Diseases 169
27. Godowski KC, Wolff ED, Thompson DM, Housley CJ, Polson AM, Dunn RL, Duke SP,
Stoller NH, Southard GL (1995) Whole mouth microbiota effects following subgingival
delivery of sanguinarium. J Periodontol 66(10):870–877
28. Görner H, Miskolczy Z, Megyesi M, Biczók L (2011) Photooxidation of alkaloids:
considerable quantum yield enhancement by rose bengal-sensitized singlet molecular
oxygen generation. Photochem Photobiol 87(6):1315–1320
29. Han MH, Kim GY, Yoo YH, Choi YH (2013) Sanguinarine induces apoptosis in human
colorectal cancer HCT-116 cells through ROS-mediated Egr-1 activation and mitochondrial
dysfunction. Toxicol Lett 220:157–166
30. Han MH, Park C, Jin CY, Kim GY, Chang YC, Moon SK, Kim WJ, Choi YH (2013)
Apoptosis induction of human bladder cancer cells by sanguinarine through reactive oxygen
species-mediated up-regulation of early growth response gene-1. PLoS ONE 8(5):e63425
31. Hazra S, Suresh Kumar G (2014) Structural and thermodynamic studies on the interaction of
iminium and alkanolamine forms of sanguinarine with hemoglobin. J Phys Chem B 118
(14):3771–3784
32. Holy J, Lamont G, Perkins E (2006) Disruption of nucleocytoplasmic trafficking of cyclin D1
and topoisomerase II by sanguinarine. BMC Cell Biol 7:13
33. Hossain M, Kabir A, Suresh Kumar G (2012) Binding of the anticancer alkaloid sanguinarine
with tRNAphe: spectroscopic and calorimetric studies. J Biomol Struct Dyn 30(2):223–234
34. Hossain M, Suresh Kumar G (2009) DNA binding of benzophenanthridine compounds
sanguinarine versus ethidium: Comparative binding and thermodynamic profile of
intercalation. J Chem Thermodyn 41(6):764–774
35. Hossain M, Khan AY, Suresh Kumar G (2011) Interaction of the anticancer plant alkaloid
sanguinarine with bovine serum albumin. PLoS ONE 6(4):e18333
36. Hossain M, Khan AY, Suresh Kumar G (2012) Study on the thermodynamics of the binding
of iminium and alkanolamine forms of the anticancer agent sanguinarine to human serum
albumin. J Chem Thermodyn 47:90–99
37. Huh J, Liepins A, Zielonka J, Andrekopoulos C, Kalyanaraman B, Sorokin A (2006)
Cyclooxygenase 2 rescues LNCaP prostate cancer cells from sanguinarine-induced apoptosis
by a mechanism involving inhibition of nitric oxide synthase activity. Cancer Res 66(7):3726–
3736
38. Jang BC, Park JG, Song DK, Baek WK, Yoo SK, Jung KH, Park GY, Lee TY, Suh SI (2009)
Sanguinarine induces apoptosis in A549 human lung cancer cells primarily via cellular
glutathione depletion. Toxicol In Vitro 23(2):281–287
39. Janovská M, Kubala M, Šimánek V, Ulrichová J (2009) Fluorescence of sanguinarine:
fundamental characteristics and analysis of interconversion between various forms. Anal
Bioanal Chem 395:235–240
40. Jash C, Payghan PV, Ghoshal N, Suresh Kumar G (2014) Binding of the iminium and
alkanolamine forms of sanguinarine to lysozyme: spectroscopic analysis, thermodynamics,
and molecular modeling studies. J Phys Chem B 118(46):13077–13091
41. Jeng JH, Wu HL, Lin BR, Lan WH, Chang HH, Ho YS, Lee PH, Wang YJ, Wang JS,
Chen YJ, Chang MC (2007) Antiplatelet effect of sanguinarine is correlated to calcium
mobilization, thromboxane and cAMP production. Atherosclerosis 191(2):250–258
42. Jones RR, Harkrader RJ, Southard GL (1986) The effect of pH on sanguinarine iminium ion
form. J Nat Prod 49(6):1109–1111
43. Kaminskyy V, Kulachkovskyy O, Stoika R (2008) A decisive role of mitochondria in defining
rate and intensity of apoptosis induction by different alkaloids. Toxicol Lett 177:168–181
44. Kantas D, Papatsiros VG, Tassis PD, Athanasiou LV, Tzika ED (2015) The effect of a natural
feed additive (Macleaya cordata), containing sanguinarine, on the performance and health
status of weaning pigs. Anim Sci J 86(1):92–98
45. Kemény-Beke A, Aradi J, Damjanovich J, Beck Z, Facskó A, Berta A, Bodnár A (2006)
Apoptotic response of uveal melanoma cells upon treatment with chelidonine, sanguinarine
and chelerythrine. Cancer Lett 237(1):67–75
170 P. Basu and G.S. Kumar
46. Kim S, Lee TJ, Leem J, Choi KS, Park JW, Kwon TK (2008) Sanguinarine-induced apoptosis:
generation of ROS, down-regulation of Bcl-2, c-FLIP, and synergy with TRAIL. J Cell
Biochem 104(3):895–907
47. Kosina P, Walterová D, Ulrichová J, Lichnovský V, Stiborová M, Rýdlová H, Vicar J,
Krecman V, Brabec MJ, Simánek V (2004) Sanguinarine and chelerythrine: assessment of
safety on pigs in ninety days feeding experiment. Food Chem Toxicol 42(1):85–91
48. Laster LL, Lobene RR (1990) New perspectives on sanguinaria clinicals: individual toothpaste
and oral rinse testing. J Can Dent Assoc 56(7):19–30
49. Lee B, Lee SJ, Park SS, Kim SK, Kim SR, Jung JH, Kim WJ, Moon SK (2008)
Sanguinarine-induced G1-phase arrest of the cell cycle results from increased p27KIP1
expression mediated via activation of the Ras/ERK signaling pathway in vascular smooth
muscle cells. Arch Biochem Biophys 471(2):224–231
50. Li W, Li H, Yao H, Mu Q, Zhao G, Li Y, Hu H, Niu X (2014) Pharmacokinetic and
anti-inflammatory effects of sanguinarine solid lipid nanoparticles. Inflammation 37(2):632–
638
51. Lopus M, Panda D (2006) The benzophenanthridine alkaloid sanguinarine perturbs
microtubule assembly dynamics through tubulin binding: a possible mechanism for its
antiproliferative activity. FEBS J 273(10):2139–2150
52. Lu JJ, Bao JL, Chen XP, Huang M, Wang YT (2012) Alkaloids isolated from natural herbs as
the anticancer agents. Evid Based Complement Altern Med. Article ID 485042
53. Mackraj I, Govender T, Gathiram P (2008) Sanguinarine. Cardiovasc Ther 26(1):75–83
54. Maiti M, Chatterjee A (1995) Production of singlet oxygen by sanguinarine and berberine.
Curr Sci 68:734–736
55. Maiti M, Das S, Sen A, Das A, Suresh Kumar G, Nandi R (2002) Influence of DNA structures
on the conversion of sanguinarine alkanolamine form to iminium form. J Biomol Struct Dyn
20(3):455–464
56. Maiti M, Nandi R, Chaudhuri K (1983) The effect of pH on the absorption and fluorescence
spectra of sanguinarine. Photochem Photobiol 38(2):245–249
57. Maiti M, Nandi R, Chaudhuri K (1982) Sanguinarine: a monofunctional intercalating alkaloid.
FEBS Lett 142(2):280–284
58. Malikova J, Zdarilova A, Hlobilkova A, Ulrichova J (2006) The effect of chelerythrine on cell
growth, apoptosis, and cell cycle in human normal and cancer cells in comparison with
sanguinarine. Cell Biol Toxicol 22:439–453
59. Matkar SS, Wrischnik LA, Hellmann-Blumberg U (2008) Production of hydrogen peroxide
and redox cycling can explain how sanguinarine and chelerythrine induce rapid apoptosis.
Biochem Biophys 477:43–52
60. Mo S, Xiong H, Shu G, Yang X, Wang J, Zheng C, Xiong W, Mei Z (2013)
Phaseoloideside E, a novel natural triterpenoid saponin identified from Entada phaseoloides,
induces apoptosis in Ec-109 esophageal cancer cells through reactive oxygen species
generation. J Pharmacol Sci 122(3):163–175
61. Nandi R, Chaudhuri K, Maiti M (1985) Effects of ionic strength and pH on the binding of
sanguinarine to deoxyribonucleic acid. Photochem Photobiol 42(5):497–503
62. Niu X, Fan T, Li W, Xing W, Huang H (2012) The anti-inflammatory effects of sanguinarine
and its modulation of inflammatory mediators from peritoneal macrophages. Eur J Pharmacol
689(1–3):262–269
63. Orvos P, Virág L, Tálosi L, Hajdú Z, Csupor D, Jedlinszki N, Szél T, Varró A, Hohmann J
(2015) Effects of Chelidonium majus extracts and major alkaloids on hERG potassium
channels and on dog cardiac action potential—a safety approach. Fitoterapia 100:156–165
64. Park H, Bergeron E, Senta H, Guillemette K, Beauvais S, Blouin R, Sirois J, Faucheux N
(2010) Sanguinarine induces apoptosis of human osteosarcoma cells through the extrinsic and
intrinsic pathways. Biochem Biophys Res Commun 399:446–451
65. Park SY, Jin ML, Kim YH, Kim CM, Lee SJ, Park G (2014) Involvement of heme oxygenase1
in neuroprotection by sanguinarine against glutamate-triggered apoptosis inHT22 neuronal
cells. Environ Toxicol Pharmacol 38(3):701–710
7 Sanguinarine and Its Role in Chronic Diseases 171
66. Parsons LG, Thomas LG, Southard GL, Woodall IR, Jones BJ (1897) Effect of sanguinaria
extract on established plaque and gingivitis when supragingivally delivered as a manual rinse
or under pressure in an oral irrigator. J Clin Periodontol 14(7):381–385
67. Philogène BJ, Arnason JT, Towers GH, Abramowski Z, Campos F, Champagne D,
McLachlan D (1984) Berberine: a naturally occurring phototoxic alkaloid. J Chem Ecol
10:115–123
68. Pickler L, Beirão BCB, Hayashi RM, Durau JF, Lourenço MC, Caron LF, Santin E (2013)
Effect of sanguinarine in drinking water on Salmonella control and the expression of immune
cells in peripheral blood and intestinal mucosa of broilers. J Appl Poult res 22:430–438
69. Pridgeon JW, Klesius PH, Dominowski PJ, Yancey RJ, Kievit MS (2013) Effects of
praziquantel and sanguinarine on expression of immune genes and susceptibility to
Aeromonas hydrophila in goldfish (Carassius auratus) infected with Dactylogyrus
intermedius. Fish Shellfish Immunol 35:1301–1308
70. Selvi BR, Pradhan SK, Shandilya J, Das C, Sailaja BS, Naga Shankar G, Gadad SS, Reddy A,
Dasgupta D, Kundu TK (2009) Sanguinarine interacts with chromatin, modulates epigenetic
modifications, and transcription in the context of chromatin. Chem Biol 16(2):203–216
71. Sen A, Maiti M (1994) Interaction of sanguinarine iminium and alkanolamine form with calf
thymus DNA. Biochem Pharmacol 48(11):2097–2102
72. Serafim TL, Matos JA, Sardão VA, Pereira GC, Branco AF, Pereira SL, Parke D, Perkins EL,
Moreno AJ, Holy J, Oliveira PJ (2008) Sanguinarine cytotoxicity on mouse melanoma
K1735-M2 cells–nuclear vs. mitochondrial effects. Biochem Pharmacol 76(11):1459–1475
73. Sharma BD, Malhotra S, Bhatia V, Rathee M (1999) Epidemic dropsy in India. Postgrad
Med J 75(889):657–661
74. Singh CK, Kaur S, George J, Nihal M, Hahn MC, Scarlett CO, Ahmad N (2015) Molecular
signatures of sanguinarine in human pancreatic cancer cells: a large scale label-free
comparative proteomics approach. Oncotarget 6(12):10335–10349
75. Suresh Kumar G, Das A, Maiti M (1997) Photochemical conversion of sanguinarine to
oxysanguinarine. J Photochem Photochem A 111:51–56
76. Suresh Kumar G, Hazra S (2014) Sanguinarine, a promising anticancer therapeutic:
photochemical and nucleic acid binding properties. RSC Adv 4:56518–56531
77. Wynn SG, Wynn DVM, Fougère B (2007) Veterinary herbal medicine. Mater Med 24:459–
672
78. Tuveson RW, Larson RA, Marley KA, Wang GR, Berenbaum MR (1989) Sanguinarine, a
phototoxic H2O2-producing alkaloid. Photochem Photobiol 50(6):733–738
79. Vogt A, Tamewitz A, Skoko J, Sikorski RP, Giuliano KA, Lazo JS (2005) The benzo[c]
phenanthridine alkaloid, sanguinarine, is a selective, cell-active inhibitor of mitogen-activated
protein kinase phosphatase-1. J Biol Chem 280(19):19078–19086
80. Vrba J, Dolezel P, Vicar J, Ulrichova J (2009) Cytotoxic activity of sanguinarine and
dihydrosanguinarine in human promyelocytic leukemia HL-60 cells. Toxicol In Vitro 23:580–
588
81. Vrba J, Hrbac J, Ulrichova J, Modriansky M (2004) Sanguinarine is a potent inhibitor of
oxidative burst in DMSO-differentiated HL-60 cells by a non-redox mechanism. Chem Biol
Interact 147:35–47
82. Wang L, Cao H, Lu N, Liu L, Wang B, Hu T, Israel DA, Peek RM Jr, Polk BD, Ya F (2013)
Berberine inhibits proliferation and down-regulates epidermal growth factor receptor through
activation of Cbl in colon tumor cells. PLoS ONE 8(2):e56666
83. Wang Q, Zhao Z, Shang J, Xia W (2014) Targets and candidate agents for type 2 diabetes
treatment with computational bioinformatics approach. J Diabetes Res 2014:763936
84. Wang XQ, Wang XM, Zhou TF, Dong LQ (2012) Screening of differentially expressed genes
and small molecule drugs of paediatric allergic asthma with DNA microarray. Eur Rev Med
Pharmacol Sci 16(14):1961–1966
85. Weerasinghe P, Hallock S, Tang SC, Liepins A (2001) Sanguinarine induces bimodal cell
death in K562 but not in high Bcl-2-expressing JM1 cells. Pathol Res Pract 197(11):717–726
172 P. Basu and G.S. Kumar
Keywords Anti-depressant effects Bioavailability enhancer Insulin-resistance
Piperine
8.1 Introduction
daily for 21 days in male Wistar rats affected by collagen-induced arthritis (CIA).
Piperine was effective in bringing significant changes on all the biochemical and
inflammatory and parameters studies, in particular piperine significantly reduced the
levels of pro-inflammatory mediators [interleukin-1β (IL-1β), tumor necrosis
factor-α (TNF-α), and prostaglandin-2 (PGE2)] and increased level of
interleukin-10 (IL-10). The protective effects of piperine against arthritis were also
evident from the decrease in arthritis scoring and bone histology. These results
clearly indicate that the protective role of piperine was mediated via its anti-oxidant
effect through the suppression of lipid peroxidation and boosting the anti-oxidant
defense system. Piperine seems to shift the balance of cytokines toward a bone
protecting pattern that acts to both lower levels of TNF-a, IL-1β, and raise the levels
of IL-10. Part of the beneficial anti-inflammatory and cartilage/bone protective
effects of piperine may be mediated through the inhibition of pro-inflammatory
cytokines.
Hepatic steatosis is due to an excess of plasma fatty acids, even if de novo lipoge-
nesis is also considered an important contributing factor. Previously published
studies showed that AMP-activated protein kinase (AMPK) is thought to regulate
hepatic lipogenic gene expression by inhibiting transcription factors [15]. The
inhibition of AMPK activates liver X receptor a (LXRa), a major regulator of
lipogenesis. In animal models with high-fat diet (HFD)-induced fatty liver, LXRa is
transcriptionally upregulated and consequently activates lipogenic target genes, thus
8 Piperine and Its Role in Chronic Diseases 177
exacerbating hepatic steatosis [16]. Piperine plays a role in the transcriptional reg-
ulation of LXRa; in particular it antagonized LXRa transcriptional activity by
abolishing the interaction of ligand-bound LXRa with the co-activator
CREB-binding protein. The effects of piperine on hepatic lipid accumulation were
likely regulated via alterations in LXRa-mediated lipogenesis in mice fed a high-fat
diet. Piperine positive effects on insulin-resistance and hepatic steatosis were
reported by Choi et al. [17]. In this study, Authors examined the effect of piperine on
hepatic steatosis and insulin-resistance induced in mice by feeding a high-fat diet
(HFD) for 13 weeks. Administration of piperine (50 mg/kg body weight) to mice
with HFD-induced hepatic steatosis resulted in a significant increase in plasma
adiponectin levels. Also, elevated plasma concentrations of insulin and glucose and
hepatic lipid levels induced by feeding a HFD were reversed in mice when they were
administered piperine. Piperine reversed HFD-induced downregulation of
adiponecitn-AMPK signaling molecules which play an important role in mediating
lipogenesis, fatty acid oxidation, and insulin signaling in the livers of mice. Piperine
significantly decreased the phosphorylation of insulin receptor substrate-1 (IRS-1)
compared with the HFD-fed mice. The positive effects of piperine were confirmed
by Rondanelli et al. [18] in humans. They administered a combination of bioactive
food ingredients (epigallocatechin gallate, capsaicins, piperine, and L-carnitine)
versus placebo, in a randomized, double-blind, 8 weeks trial, involving 86 over-
weight subjects. Consumption of the dietary supplement was associated with a
significantly greater decrease in insulin-resistance, assessed by homeostasis model
assessment, leptin/adiponectin ratio, respiratory quotient, LDL-cholesterol. Leptin,
ghrelin, C-reactive protein decreased, and resting energy expenditure increased
significantly in the supplemented group compared to placebo. These results suggest
that piperine, in combination with epigallocatechin gallate, capsaicins, and
L-carnitine, could be useful for the treatment of obesity-related inflammatory
metabolic dysfunctions.
In literature there are studies about the possible analgesic, and anti-pyretic effects of
piperine as reported by Evan Prince et al. [22]. Authors treated mice with piperine
(20 and 30 mg/kg) intraperitoneally; hot plate reaction test and acetic acid test were
used to determine the analgesic activity of piperine in mice. It was found that
piperine has significant analgesic and anti-pyretic activities without ulcerogenic
effects. The results were comparable with indomethacin which was used as standard
drug for reference. Despite that, further studies are required to elucidate the
mechanism of piperine to confirm these activities.
effects alone. These results suggest that piperine enhances the neuroprotective
effects of quercitin against chronic unpredictable stress-induced oxidative stress,
neuro-inflammation and memory deficits.
Piperine also enhanced phyllanthin bioavailability as published by Sethiya et al.
[41]. These Authors evaluated the hepato-protective effects of phyllanthin along
with piperine in a mixed micellar lipid formulation (MMLF). Authors compared
phyllanthin (30 mg/kg p.o.), a complex phosphatidylcholine formulation of phyl-
lanthin (CP–PC) (30 mg/kg p.o.), phyllanthin + piperine (CP–P–PC) (30 mg/kg p.
o.), and the reference drug silymarin (100 mg/kg, p.o.) administered daily to rats for
10 days, followed by liver damage by administering a 1:1 (v/v) mixture of CCl4
and olive oil (1 ml/kg, i.p.) for 7 days from day 4 to day 10. The degree of
protection was evaluated by determining the level of marker enzymes (SGOT and
SGPT), bilirubin, and total proteins.
CP–P–PC (30 mg/kg p.o.) showed significant hepato-protective effect by
reducing the levels of serum marker enzymes (SGOT, SGPT, and bilirubin),
whereas, elevated the levels of depleted total protein, lipid peroxidation and
anti-oxidant marker enzyme activities such as, glutathione, superoxide dismutase,
catalase, glutathione peroxidase, and glutathione reductase. The complex MMLF
normalized adverse conditions of rat livers more efficiently than the non-formulated
phyllanthin, confirming, again, the effects of piperine in enhancing low bioavail-
ability of phyllanthin.
Piperine can be used also in combination with resveratrol to enhance its
bioavailability [42]: in this study Authors evaluated if co-supplementation of
piperine with resveratrol affects the bioavailability and efficacy of resveratrol with
regard to cognition and cerebral blood flow. In this randomized, double-blind,
placebo-controlled, within-subjects study, 23 adults were given placebo,
trans-resveratrol (250 mg) and trans-resveratrol with 20 mg piperine on separate
days at least a week apart. The results indicated that when co-supplemented,
piperine, and resveratrol significantly augmented cerebral blood flow during task
performance in comparison with placebo and resveratrol alone. Cognitive function,
mood, and blood pressure were not affected.
8.3 Conclusions
In conclusion, piperine has several important roles that can be useful in clinical
practice, in particular for reduction of insulin-resistance, inflammation, and hepatic
steatosis. However, the most far-reaching attribute of piperine is its inhibitory effect
on enzymatic drug biotransformation reactions in the liver. Piperine strongly
inhibits hepatic and intestinal aryl hydrocarbon hydroxylase and UDP-glucuronyl
transferase, thus enhancing the bioavailability of a number of therapeutic drugs as
well as phytochemicals. Piperine’s bioavailability-enhancing property is also partly
attributed to increased absorption as a result of its effect on the ultrastructure of
182 G. Derosa et al.
intestinal brush border. This promising effect of piperine has been extensively
employed to increase the bioavailability and pharmacological effects of several
drugs and phytopharmaceuticals.
References
1. Manske RHF (1965) The alkaloids. Chemistry and physiology, vol VIII. Academic Press,
New York, p 673
2. Selvendiran K, Sakthisekaran D (2004) Chemopreventive effect of piperine on modulating
lipid peroxidation and membrane bound enzymes in benzo(a)pyrene induced lung
carcinogenesis. Biomed Pharmacother 58:264–267
3. McNamara FN, Randall A, Gunthorpe MJ (2005) Effects of piperine, the pungent component
of black pepper, at the human vanilloid receptor (TRPV1). Br J Pharmacol 144(6):781–790
4. Schöffmann A, Wimmer L, Goldman D et al (2014) Efficient modulation of γ-aminobutyric
acid type A receptors by piperine derivatives. J Med Chem 57(37):5602–5619
5. Lee SA, Hong SS, Han XH et al (2005) Piperine from the fruits of Piper longum with
inhibitory effect on monoamine oxidase and antidepressant-like activity. Chem Pharm Bull
(Tokyo) 53(7):832–835
6. Murray M (2012) Toxicological actions of plant-derived and anthropogenic
methylenedioxyphenyl-substituted chemicals in mammals and insects. J Toxicol Environ
Health B Crit Rev 15(6):365–395
7. Najar IA, Sachin BS, Sharma SC et al (2010) Modulation of P-glycoprotein ATPase activity
by some phytoconstituents. Phytother Res 24(3):454–458
8. Vaibhav K, Shrivastava P, Javed H, Khan A, Ahmed ME, Tabassum R, Khan MM,
Khuwaja G, Islam F, Siddiqui MS, Safhi MM, Islam F (2012) Piperine suppresses cerebral
ischemia-reperfusion-induced inflammation through the repression of COX-2, NOS-2, and
NF-κB in middle cerebral artery occlusion rat model. Mol Cell Biochem 367(1–2):73–84
9. Srinivasan K (2007) Black pepper and its pungent principle-piperine: a review of diverse
physiological effects. Crit Rev Food Sci Nutr 47(8):735–748
10. Umar S, Golam Sarwar AH, Umar K, Ahmad N, Sajad M, Ahmad S, Katiyar CK, Khan HA
(2013) Piperine ameliorates oxidative stress, inflammation and histological outcome in
collagen induced arthritis. Cell Immunol 284(1–2):51–59
11. Toyoda T, Shi L, Takasu S, Cho YM, Kiriyama Y, Nishikawa A, Ogawa K, Tatematsu M,
Tsukamoto T (2015) Anti-Inflammatory effects of capsaicin and piperine on helicobacter
pylori-induced chronic gastritis in mongolian gerbils. Helicobacter. doi:10.1111/hel.12243
12. Cai XZ, Huang WY, Qiao Y, Du SY, Chen Y, Chen D, Yu S, Che RC, Liu N, Jiang Y (2013)
Inhibitory effects of curcumin on gastric cancer cells: a proteomic study of molecular targets.
Phytomedicine 20:495–505
13. Xia Y, Khoi PN, Yoon HJ, Lian S, Joo YE, Chay KO, Kim KK, Jung YD (2015) Piperine
inhibits IL-1b-induced IL-6 expression by suppressing p38 MAPK and STAT3 activation in
gastric cancer cells. Mol Cell Biochem 398:147–156
14. Doucette CD, Rodgers G, Liwski RS, Hoskin DW (2015) Piperine from black pepper inhibits
activation-induced proliferation and effector function of T lymphocytes. J Cell Biochem 116
(11):2577–2588
15. Nafisi S, Adelzadeh M, Norouzi Z, Sarbolouki MN (2009) Curcumin binding to DNA and
RNA. DNA Cell Biol 28:201–208
16. Hwahng SH, Ki SH, Bae EJ, Kim HE, Kim SG (2009) Role of adenosine monophosphate
activated protein kinase-p70 ribosomal S6 kinase-1 pathway in repression of liver X
receptor-alpha-dependent lipogenic gene induction and hepatic steatosis by a novel class of
dithiolethiones. Hepatology 49:1913–1925
8 Piperine and Its Role in Chronic Diseases 183
17. Choi S, Choi Y, Choi Y, Kim S, Jang J, Park T (2013) Piperine reverses high fat diet-induced
hepatic steatosis and insulin resistance in mice. Food Chem 141(4):3627–3635
18. Rondanelli M, Opizzi A, Perna S, Faliva M, Solerte SB, Fioravanti M, Klersy C, Cava E,
Paolini M, Scavone L, Ceccarelli P, Castellaneta E, Savina C, Donini LM (2013) Improvement
in insulin resistance and favourable changes in plasma inflammatory adipokines after weight
loss associated with two months’ consumption of a combination of bioactive food ingredients
in overweight subjects. Endocrine 44(2):391–401
19. Li S, Wang C, Wang M, Li W, Matsumoto K, Tang Y (2007) Antidepressant like effects of
piperine in chronic mild stress treated mice and its possible mechanisms. Life Sci 80
(15):1373–1381
20. Kirschenbaum B, Goldman SA (1995) Brain-derived neurotrophic factor promotes the
survival of neurons arising from the adult rat forebrain subependymal zone. Proc Natl Acad
Sci USA 92(1):210–214
21. Mao QQ, Huang Z, Zhong XM, Xian YF, Ip SP (2014) Piperine reverses chronic
unpredictable mild stress-induced behavioral and biochemical alterations in rats. Cell Mol
Neurobiol 34(3):403–408
22. Evan Prince S, Aayesha N, Mahima V, Mahaboobkhan R (2013) Analgesic, antipyretic and
ulcerogenic effects of piperine: an active ingredient of pepper. J Pharm Sci Res 5(10):203–206
23. Motterlini R, Foresti R, Bassi R, Green CJ (2000) Curcumin, an antioxidant and
anti-inflammatory agent, induces heme oxygenase-1 and protects endothelial cells against
oxidative stress. Free Radic Biol Med 28:1303–1312
24. Wahlstrom B, Blennow GA (1978) Study on the fate of curcumin in the rat. Acta Pharm
Toxicol 43:86–92
25. Sahebkar A, Chew GT, Watts GF (2014) Recent advances in pharmacotherapy for
hypertriglyceridemia. Prog Lipid Res 56:47–66
26. Panahi Y, Badeli R, Karami GR, Sahebkar A (2015) Investigation of the efficacy of adjunctive
therapy with bioavailability-boosted curcuminoids in major depressive disorder. Phytother Res
29(1):17–21
27. Panahi Y, Rahimnia AR, Sharafi M, Alishiri G, Saburi A, Sahebkar A (2014) Curcuminoid
treatment for knee osteoarthritis: a randomized double-blind placebo-controlled trial.
Phytother Res 28(11):1625–1631
28. Panahi Y, Saadat A, Beiraghdar F, Sahebkar A (2014) Adjuvant therapy with
bioavailability-boosted curcuminoids suppresses systemic inflammation and improves
quality of life in patients with solid tumors: a randomized double-blind placebo-controlled
trial. Phytother Res 28(10):1461–1467
29. Sahebkar A (2014) Are curcuminoids effective C-reactive protein-lowering agents in clinical
practice? Evidence from a meta-analysis. Phytother Res 28(5):633–642
30. Sahebkar A, Mohammadi A, Atabati A, Rahiman S, Tavallaie S, Iranshahi M, Akhlaghi S,
Ferns GA, Ghayour-Mobarhan M (2013) Curcuminoids modulate pro-oxidant-antioxidant
balance but not the immune response to heat shock protein 27 and oxidized LDL in obese
individuals. Phytother Res 27:1883–1888
31. Mohammadi A, Sahebkar A, Iranshahi M, Amini M, Khojasteh R, Ghayour-Mobarhan M,
Ferns GA (2013) Effects of supplementation with curcuminoids on dyslipidemia in obese
patients: a randomized crossover trial. Phytother Res 27(3):374–379
32. Panahi Y, Ghanei M, Bashiri S, Hajihashemi A, Sahebkar A (2015) Short-term curcuminoid
supplementation for chronic pulmonary complications due to sulfur mustard intoxication:
Positive results of a randomized double-blind placebo-controlled trial. Drug Res (Stuttg) 65
(11):567–573
33. Rinwa P, Kumar A (2012) Piperine potentiates the protective effects of curcumin against
chronic unpredictable stress-induced cognitive impairment and oxidative damage in mice.
Brain Res 1488:38–50
34. Banji D, Banji OJ, Dasaroju S, Annamalai AR (2013) Piperine and curcumin exhibit
synergism in attenuating D-galactose induced senescence in rats. Eur J Pharmacol 703(1–
3):91–99
184 G. Derosa et al.
35. Sehgal A, Kumar M, Jain M, Dhawan DK (2011) Combined effects of curcumin and piperine
in ameliorating benzo(a)pyrene induced DNA damage. Food Chem Toxicol 49(11):
3002–3006
36. Panahi Y, Hosseini MS, Khalili N, Naimi E, Majeed M, Sahebkar A (2015) Antioxidant and
anti-inflammatory effects of curcuminoid-piperine combination in subjects with metabolic
syndrome: A randomized controlled trial and an updated meta-analysis. Clin Nutr. doi:10.
1016/j.clnu.2014.12.019
37. Atal CK, Dubey RK, Singh J (1985) Biochemical basis of enhanced drug bioavailability by
piperine: evidence that piperine is a potent inhibitor of drug metabolism. J Pharmacol Exp
Ther 232:258–262
38. Panahi Y, Khalili N, Hosseini MS, Abbasinazari M, Sahebkar A (2014) Lipid-modifying
effects of adjunctive therapy with curcuminoids-piperine combination in patients with
metabolic syndrome: results of a randomized controlled trial. Complement Ther Med 22
(5):851–857
39. Neyrinck AM, Alligier M, Memvanga PB, Névraumont E, Larondelle Y, Préat V, Cani PD,
Delzenne NM (2013) Curcuma longa extract associated with white pepper lessens high fat
diet-induced inflammation in subcutaneous adipose tissue. PLoS ONE 8(11):e81252
40. Rinwa P, Kumar A (2013) Quercetin along with piperine prevents cognitive dysfunction,
oxidative stress and neuro-inflammation associated with mouse model of chronic
unpredictable stress. Arch Pharm Res. doi:10.1007/s12272-013-0205-4
41. Sethiya NK, Shah P, Rajpara A, Nagar PA, Mishra SH (2015) Antioxidant and
hepatoprotective effects of mixed micellar lipid formulation of phyllanthin and piperine in
carbon tetrachloride-induced liver injury in rodents. Food Funct. doi:10.1039/c5fo00947b
42. Wightman EL, Reay JL, Haskell CF, Williamson G, Dew TP, Kennedy DO (2014) Effects of
resveratrol alone or in combination with piperine on cerebral blood flow parameters and
cognitive performance in human subjects: a randomised, double-blind, placebo-controlled,
cross-over investigation. Br J Nutr 112(2):203–213
Chapter 9
Therapeutic Potential and Molecular
Targets of Piceatannol in Chronic Diseases
9.1 Introduction
Anti-aging
Anti-inflammatory
activity Immune Function
Obesity Diabetes
Piceatannol
Cadiovascular Neurodegenerative
Disease disorders
Osteoporosis Cancer
non-receptor spleen tyrosine kinase (Syk) which plays a critical role in the regulation
of not only immune and inflammatory responses of hematopoietic cells, but also
general physiological functions in a wide variety of non-hematopoietic cells [1].
As a hydroxylated analog and a metabolite of resveratrol (3,5,4′-trihydroxy-
trans-stilbene), PIC has multiple biological functions, including antioxidative,
anti-inflammatory, anticancer, antidiabetic, cardioprotective, neuroprotective, and
immunomodulatory properties (Fig. 9.1). PIC has structural similarity to resveratrol
and both compounds share a spectrum of biological activities [2, 3]. This review
summarizes the updated research findings regarding the cancer chemopreventive/
anticarcinogenic and other health promoting effects of PIC and underlying
mechanisms.
Piceatannol Antiangiogenesis
Treatment of the human colon carcinoma Caco-2 cell line with PIC resulted in the
reduced cell proliferation without influencing the differentiation [6]. Flow cyto-
metric analysis of cell cycle distribution revealed an accumulation of cells in the S
phase. Immunoblot analysis demonstrated that cyclin-dependent kinases (CDK) 2
and 6 as well as cdc2 were expressed at steady-state levels, whereas the expression
of cyclin D1, cyclin B1, and CDK 4 was downregulated. The level of p27KIP1 was
also reduced, whereas that of cyclin E was enhanced. Similar changes were also
observed in studies with HCT-116 colon cancer cells [6].
PIC also inhibits cell cycle progression in the DU145 human prostate cancer cell
line. PIC treatment enhanced the percentage of cells in the G1 phase and decreased
[3H]thymidine incorporation as well as expression of cyclin A, cyclin D1, CDK2,
and CDK4. PIC suppressed CDK4 and CDK2 activities as well, but had no effect
on the levels of p21WAF1/CIP1 or p27KIP1 [7]. PIC inhibited the proliferation of T24
and HT1376 human bladder cancer cells by blocking cell cycle progression in the
G0/G1 phase and induced apoptosis. The G0/G1 phase arrest was attributed to an
increased expression of p21WAF1/CIP1. An enhancement in Fas/APO-1 and
membrane-bound Fas ligand might be responsible for the apoptotic effect induced
by PIC in the aforementioned bladder cancer cells [8].
Leukemia
PIC and other stilbenes were investigated for their tumor-specific cytotoxicity and
apoptosis-inducing activity, using four human tumor cell lines (squamous cell
carcinoma HSC-2, HSC-3, submandibular gland carcinoma HSG and promyelo-
cytic leukemia HL-60) and three normal human oral cell lines (gingival fibroblast
HGF, pulp cell HPC, periodontal ligament fibroblast HPLF). Among the seven cell
lines, HL-60 and HSC-2 cells were the most sensitive to the cytotoxic action of
these compounds. PIC induced internucleosomal DNA fragmentation and activa-
tion of caspases-3, -8, and -9 in HL-60 cells. [13].
PIC treatment to the human leukemia cell line U937 induced the formation of
apoptotic bodies, DNA fragmentation, and the accumulation of the sub-G1 phase
[14]. The proapoptotic effect of PIC was associated with downregulation of
anti-apoptotic Bcl-2 and cIAP-2, proteolytic activation of caspase-3, and the
degradation/cleavage of PARP. z-DEVD-fmk, a caspase-3-specific inhibitor,
attenuated the proapoptotic activity of PIC in U937 cells [14]. PIC-induced apop-
totic death of human leukemia U937 cells was accompanied by an increase in the
intracellular Ca2+ concentration ([Ca2+]i), inactivation of extracellular signal-
regulated kinase (ERK), activation of p38 mitogen-activated protein kinase
(MAPK), degradation of procaspase-8 and production of t-Bid [15]. PIC treatment
increased the levels of Fas and FasL and their mRNA transcripts. Downregulation
of FADD blocked PIC-induced procaspase-8 degradation and rescued viability of
9 Therapeutic Potential and Molecular Targets of Piceatannol … 189
PIC inhibited the growth of the androgen receptor-negative prostate PC3 cancer cell
line which was associated with accumulation of endogenous ceramide [17]. In
another study, the effects of PIC on proliferation of androgen-dependent LNCaP
and androgen-independent DU145 and PC-3 prostate cancer cells were investigated
[18]. PIC exerted anti-proliferative effects, which was accompanied by cell cycle
blockade in G1/S and S phases in LNCaP and PC-3 cells and induction of apoptosis
in DU145 cells [18]. PIC induced apoptosis of DU145 cells was evidenced by
cleavage of caspase 3 and apoptosis inducing factor AIF and an increase in total cyt
c. The apoptotic changes occurred in concordance with DNA damage, as revealed
by increased phosphorylation of histone H2AX at Ser139. Treatment of
different-stage prostate cancer cells with PIC also resulted in cell type-specific
downregulation of the mammalian target of rapamycin (mTOR), a kinase involved
in growth control of eukaryotic cells [18]. PIC also increased the protein levels of
cleaved caspase-8, -9, -7, and -3 and cleaved PARP in DU145 cells, thereby
inducing apoptosis [19]. PIC induced mitochondrial outer membrane permeability
changes, resulting in cyt c release from the mitochondria to the cytosol. PIC
treatment enhanced the levels of truncated Bid, Bax, Bik, and Bok with a con-
comitant decrease in the levels of Mcl-1 and Bcl-xL [19]. These results indicate that
PIC induces apoptosis via the activation of the death receptor and mitochondrial-
dependent pathways in prostate cancer cells.
PIC induced HCT116 and HT29 colon cancer cell apoptosis by promoting
expression of microRNA-129, and suppressing expression of Bcl-2, a known target
of microRNA-129. Moreover, knock down of microRNA-129 reversed the
reduction of cell viability induced by PIC in these cells [20]. PIC exerted
anti-proliferative activities in prostate cancer cell lines. The anti-tumorigenic effects
of PIC was determined in LNCaP-Luc-xenografts. Two-week pretreatment with
PIC diminished cell colonization, tumor volume, and tumor growth in the xeno-
grafts [21]. Treatment of MDA-MB-231 human breast cancer cells with PIC caused
a rapid release of calcium from the endoplasmic reticulum. This elevation of
intracellular calcium modulates the activity of p53 and the subsequent transcription
of several genes encoding proapoptotic proteins [22]. Nanomolar concentration of
PIC induced c-Myc expression and activated survival signaling in MCF-7 human
breast cancer cells, leading to accelerated cell proliferation [23]. In contrast, PIC at
the concentrations in the micromolar range suppressed c-Myc expression and
induced apoptosis.
190 Y.-J. Surh and H.-K. Na
Environmental conditions or chemical agents can interfere with the function of the
endoplasmic reticulum (ER), and the resultant ER stress can be toxic to the cell if it
is not relieved. Agents that cause ER stress in cancer cells can elicit anticancer
activity. PIC was identified as one of the potent inducers of ER stress. In the
HT1080 reporter cells treated with PIC, there were both splicing of XBP1 and
induction of the ATF4 target gene product CCAAT-enhancer-binding protein
homologous protein (CHOP), leading to ER stress [24]. This was accompanied by a
modest increase in LC3 processing, indicative of a mild autophagic response.
The health beneficial effect of resveratrol may be associated, at least in part, with
its capacity to promote autophagy by activating the NAD+-dependent deacetylase
sirtuin 1 [25]. PIC is a weak activator of sirtuin 1, and its induction of autophage is
independent of this enzyme. Resveratrol and PIC, in combination, induced autop-
hagy and stimulated the deacetylation of cytoplasmic proteins more efficiently than
either compound alone. These findings suggest that both compounds, through
different mechanisms, may synergize in inducing autophagy [25].
Cancer invasion and metastasis are two main causes of treatment failure and
cancer-related mortality. Several studies have demonstrated that matrix metallo-
proteinases (MMPs) are critical for tumor invasion and metastasis. PIC inhibited
MMP-2 activity and manifestation of the invasive phenotype of MCF10A human
breast epithelial cells harboring mutated H-ras (H-ras MCF10A cells) more
effectively than resveratrol [27]. PIC attenuated the H-ras-induced phosphorylation
9 Therapeutic Potential and Molecular Targets of Piceatannol … 191
agreement with the observation in the previous study by Minakawa et al. [45], a
single intragastric administration of PIC reduced the blood glucose levels in db/db
mice [46]. In skeletal muscle cells, glucose uptake occurs mainly through glucose
transporter 4 (GLUT4) in response to insulin. 5′-Adenosine monophosphate-
activated protein kinase (AMPK) is known as a GLUT4 translocation promoter, and
AMPK activators, hence have a potential to overcome insulin resistance in the
diabetic state. PIC increased glucose uptake in L6 myotubes by promoting GLUT4
translocation to plasma membrane via AMPK activation [45].
The hypoglycemic effect of PIC was examined after intravascular administration
to healthy rats. Intravascularly administered PIC reduced the blood glucose con-
centrations during both fasting and the glucose tolerance test [47]. Furthermore,
PIC increased the insulinogenic index during glucose tolerance tests but had no
influence on insulin sensitivity. Based on these observations, it is likely that PIC
given orally enhances glucose tolerance, and this effect is mediated by intact PIC
itself, not its metabolite, by stimulating early phase secretion of insulin.
PIC effectively mitigated the inhibitory effects of palmitic acid on the
insulin-mediated phosphorylation of insulin receptor substrate-1 in human umbil-
ical vein endothelial cells (HUVECs). The protective effect of PIC against palmitic
acid-induced impairment of insulin signaling was mediated by heme oxygenase-1
(HO-1) activity [48].
ROS accumulation were observed after PIC treatment to PC12 cells [52]. PIC
significantly promoted activation of anti-apoptotic Bad and Akt through phos-
phorylation, while it reduced the Bcl-2/Bax ratio and cleavage of caspase-9,
caspase-3, and PARP.
Inflammation occurs in the ischemic brain, which consists of various types of cells,
such as neurons, endothelial cells, and immune cells. Macrophage-inducible C-type
lectin (Mincle, CLEC4E) receptor is involved in neuroinflammation in cerebral
ischemia and traumatic brain injury. Mincle, its ligand SAP130, and its downstream
tyrosine kinase Syk were upregulated in the brain after ischemia, and participate in
the pathogenesis of ischemic stroke by initiating inflammation [1]. In mice treated
with PIC intraperitoneally before ischemia and just after reperfusion, the cerebral
ischemic injury was ameliorated. The protective effects of PIC against ischemic
brain injury were attributed to its inhibition of Syk [1]. Subarachnoid hemorrhage
(SAH) accounts for 5–7 % of all strokes. PIC reduced brain edema and ameliorated
neurological deficits after SAH in a rat stroke model [54].
9.3.3.4 Neuroinflammation
Mast cells and basophils are major effector cells in the immunoglobulin E (IgE)-
dependent allergic reactions as well as in the innate immunity. Upon allergen
exposure, these cells are stimulated via the high affinity IgE receptor (FcepsilonRI)
to release several proinflammatory chemical mediators, such as leukotrienes,
immunoregulatory cytokines, and histamine. FcepsilonRI-mediated signaling is
initiated upon tyrosine phosphorylation of this receptor by the Src family kinase
Lyn, which is followed by an activation of Syk. PIC, as a classical Syk kinase
inhibitor, can modulate the FcepsilonRI-mediated signaling in mast cells, and has a
potential for use in the treatment of inflammatory and allergy diseases [67].
9 Therapeutic Potential and Molecular Targets of Piceatannol … 199
Histamine is one of the key chemical mediators that play an important role in
allergic inflammation. PIC 4′-b-glucoside formed in cultured Phytolacca
Americana treated with PIC showed the strong inhibitory effects on histamine
release from rat peritoneal mast cells [68]. Compound 48/80 (8 mg/kg) was used to
induce a systemic fatal allergic reaction in mice. Intraperitoneal administration of
PIC inhibited the compound 48/80-induced systemic anaphylaxis and serum his-
tamine release [69]. PIC treatment also ameliorated local allergic reactions in an
IgE-mediated passive cutaneous anaphylaxis model [70]. The rat basophilic leu-
kemia RBL-2H3 cell line exhibits phenotypic characteristics of mucosal mast cells.
After stimulation with antigens cells release histamine and b-hexosaminidase, a
marker of mast cell degranulation. PIC pretreatment strongly suppressed histamine
release and degranulation in the RBL-2H3 cells stimulated with phorbol
12-myristate 13-acetate (PMA) and the calcium ionophore A23187 [69]. PIC
treatment of the human mast cell line HMC-1 reduced PMA plus A23187-induced
mRNA expression and release of the proinflammatory cytokines (TNF-a and IL-8).
9.3.9.2 Melanogenesis
The effects of passion fruit on melanin inhibition and collagen synthesis were
studied using cultured human melanoma and fibroblast cells [80]. Treatment of
melanoma cells with the seed part of passion fruit resulted in inhibition of
melanogenesis. The production of total soluble collagen in cultured dermal
fibroblast cells was also elevated. PIC is speculated as the major component
responsible for the passion fruit effects on melanogenesis and collagen synthesis
[80]. The skin lightening potential of PIC was investigated in terms of its ability to
inhibit melanogenesis [81]. PIC strongly inhibited tyrosinase activity involved in
melanin biosynthesis and lowered the melanin content in cultured B16F10 mela-
noma cells. The antioxidative property is closely linked to the melanogenic activity.
PIC reduced the ROS accumulation and the ratio of GSH to its oxidized form
GSSG in these cells [81].
Inflammatory response has a critical role in neuronal damage after retinal I/R injury,
and is regulated tightly by TLR4. The expression of TLR4 was upregulated after
I/R [89]. TLR4 knockout (KO) mice were much less susceptible to the histologic
damage causedby I/R compared to wild-type mice. The phosphorylation level of
NF-jB after I/R in TLR4 KO mice was decreased compared to that in wild-type
mice. The expression of phosphorylated Syk was upregulated after I/R, and this was
blunted in TLR4 KO mice. PIC inhibited the histologic and functional retinal
damage, and reduced the phosphorylation level of NF-jB induced by I/R [89].
Pre- or post-treatment of PIC significantly blocked the LPS-induced ocular
inflammation in rats [90]. Further, PIC also suppressed the expression of COX-2
and iNOS and activation of NF-jB in the ciliary bodies as well as retina, and also in
primary human non-pigmented ciliary epithelial cells treated with LPS. Similarly,
PIC diminished the LPS-induced production of NO and PGE2 in these cells [90].
human microsomal
CYP1B1 and CYP1A1/2
bacterial CYP102A1
Resveratrol Piceatannol
Fig. 9.3 Formation of PIC from resveratrol by human microsomal/recombinant CYP1B1 and
CYP1A1/2 activities. The bacterial CYP101A1 also catalyzes the formation of PIC
204 Y.-J. Surh and H.-K. Na
References
10. Schmeel FC, Schmeel LC, Kim Y, Schmidt-Wolf IG (2014) Piceatannol exhibits selective
toxicity to multiple myeloma cells and influences the Wnt/beta-catenin pathway. Hematol
Oncol 32:197–204
11. Schmeel LC, Schmeel FC, Kim Y, Endo T, Lu D, Schmidt-Wolf IG (2013) Targeting the
Wnt/beta-catenin pathway in multiple myeloma. Anticancer Res 33:4719–4726
12. Wieder T, Prokop A, Bagci B, Essmann F, Bernicke D, Schulze-Osthoff K, Dorken B,
Schmalz HG, Daniel PT, Henze G (2001) Piceatannol, a hydroxylated analog of the
chemopreventive agent resveratrol, is a potent inducer of apoptosis in the lymphoma cell line
BJAB and in primary, leukemic lymphoblasts. Leukemia 15:1735–1742
13. Chowdhury SA, Kishino K, Satoh R, Hashimoto K, Kikuchi H, Nishikawa H, Shirataki Y,
Sakagami H (2005) Tumor-specificity and apoptosis-inducing activity of stilbenes and
flavonoids. Anticancer Res 25:2055–2063
14. Kim YH, Park C, Lee JO, Kim GY, Lee WH, Choi YH, Ryu CH (2008) Induction of
apoptosis by piceatannol in human leukemic U937 cells through down-regulation of Bcl-2
and activation of caspases. Oncol Rep 19:961–967
15. Liu WH, Chang LS (2010) Piceatannol induces Fas and FasL up-regulation in human
leukemia U937 cells via Ca2+/p38alpha MAPK-mediated activation of c-Jun and ATF-2
pathways. Int J Biochem Cell Biol 42:1498–1506
16. Kang CH, Moon DO, Choi YH, Choi IW, Moon SK, Kim WJ, Kim GY (2011) Piceatannol
enhances TRAIL-induced apoptosis in human leukemia THP-1 cells through Sp1- and
ERK-dependent DR5 up-regulation. Toxicol In Vitro 25:605–612
17. Sala G, Minutolo F, Macchia M, Sacchi N, Ghidoni R (2003) Resveratrol structure and
ceramide-associated growth inhibition in prostate cancer cells. Drugs Exp Clin Res 29:263–
269
18. Hsieh TC, Lin CY, Lin HY, Wu JM (2012) AKT/mTOR as Novel targets of polyphenol
piceatannol possibly contributing to inhibition of proliferation of cultured prostate cancer
cells. ISRN Urol 2012:272697
19. Kim EJ, Park H, Park SY, Jun JG, Park JH (2009) The grape component piceatannol induces
apoptosis in DU145 human prostate cancer cells via the activation of extrinsic and intrinsic
pathways. J Med Food 12:943–951
20. Zhang H, Jia R, Wang C, Hu T, Wang F (2014) Piceatannol promotes apoptosis via
up-regulation of microRNA-129 expression in colorectal cancer cell lines. Biochem Biophys
Res Commun 452:775–781
21. Dias SJ, Li K, Rimando AM, Dhar S, Mizuno CS, Penman AD, Levenson AS (2013)
Trimethoxy-resveratrol and piceatannol administered orally suppress and inhibit tumor
formation and growth in prostate cancer xenografts. Prostate 73:1135–1146
22. van Ginkel PR, Yan MB, Bhattacharya S, Polans AS, Kenealey JD (2015) Natural products
induce a G protein-mediated calcium pathway activating p53 in cancer cells. Toxicol Appl
Pharmacol 288:453–462
23. Vo NT, Madlener S, Bago-Horvath Z, Herbacek I, Stark N, Gridling M, Probst P,
Giessrigl B, Bauer S, Vonach C, Saiko P, Grusch M, Szekeres T, Fritzer-Szekeres M,
Jager W, Krupitza G, Soleiman A (2010) Pro- and anticarcinogenic mechanisms of
piceatannol are activated dose dependently in MCF-7 breast cancer cells. Carcinogenesis
31:2074–2081
24. Papandreou I, Verras M, McNeil B, Koong AC, Denko NC (2015) Plant stilbenes induce
endoplasmic reticulum stress and their anti-cancer activity can be enhanced by inhibitors of
autophagy. Exp Cell Res 339:147–153
25. Pietrocola F, Marino G, Lissa D, Vacchelli E, Malik SA, Niso-Santano M, Zamzami N,
Galluzzi L, Maiuri MC, Kroemer G (2012) Pro-autophagic polyphenols reduce the
acetylation of cytoplasmic proteins. Cell Cycle 11:3851–3860
26. Azmi AS, Bhat SH, Hadi SM (2005) Resveratrol-Cu(II) induced DNA breakage in human
peripheral lymphocytes: implications for anticancer properties. FEBS Lett 579:3131–3135
9 Therapeutic Potential and Molecular Targets of Piceatannol … 207
27. Song NR, Hwang MK, Heo YS, Lee KW, Lee HJ (2013) Piceatannol suppresses the
metastatic potential of MCF10A human breast epithelial cells harboring mutated H-ras by
inhibiting MMP-2 expression. Int J Mol Med 32:775–784
28. Ko HS, Lee HJ, Kim SH, Lee EO (2012) Piceatannol suppresses breast cancer cell invasion
through the inhibition of MMP-9: involvement of PI3K/AKT and NF-kappaB pathways.
J Agric Food Chem 60:4083–4089
29. Kwon GT, Jung JI, Song HR, Woo EY, Jun JG, Kim JK, Her S, Park JH (2012) Piceatannol
inhibits migration and invasion of prostate cancer cells: possible mediation by decreased
interleukin-6 signaling. J Nutr Biochem 23:228–238
30. Jayasooriya RG, Lee YG, Kang CH, Lee KT, Choi YH, Park SY, Hwang JK, Kim GY
(2013) Piceatannol inhibits MMP-9-dependent invasion of tumor necrosis
factor-alpha-stimulated DU145 cells by suppressing the Akt-mediated nuclear
factor-kappaB pathway. Oncol Lett 5:341–347
31. Kita Y, Miura Y, Yagasaki K (2012) Antiproliferative and anti-invasive effect of piceatannol,
a polyphenol present in grapes and wine, against hepatoma AH109A cells. J Biomed
Biotechnol 2012:672416
32. Wesolowska O, Wisniewski J, Duarte N, Ferreira MJ, Michalak K (2007) Inhibition of
MRP1 transport activity by phenolic and terpenic compounds isolated from Euphorbia
species. Anticancer Res 27:4127–4133
33. Borge M, Remes Lenicov F, Nannini PR, de los Rios Alicandu MM, Podaza E, Ceballos A,
Fernandez Grecco H, Cabrejo M, Bezares RF, Morande PE, Oppezzo P, Giordano M,
Gamberale R (2014) The expression of sphingosine-1 phosphate receptor-1 in chronic
lymphocytic leukemia cells is impaired by tumor microenvironmental signals and enhanced
by piceatannol and R406. J Immunol 193:3165–3174
34. Song H, Jung JI, Cho HJ, Her S, Kwon SH, Yu R, Kang YH, Lee KW, Park JH (2015)
Inhibition of tumor progression by oral piceatannol in mouse 4T1 mammary cancer is
associated with decreased angiogenesis and macrophage infiltration. J Nutr Biochem
26:1368–1378
35. Xu B, Tao ZZ (2015) Piceatannol enhances the antitumor efficacy of gemcitabine in human
A549 non-small cell lung cancer cells. Oncol Res 22:213–217
36. Farrand L, Byun S, Kim JY, Im-Aram A, Lee J, Lim S, Lee KW, Suh JY, Lee HJ, Tsang BK
(2013) Piceatannol enhances cisplatin sensitivity in ovarian cancer via modulation of p53,
X-linked inhibitor of apoptosis protein (XIAP), and mitochondrial fission. J Biol Chem
288:23740–23750
37. Fritzer-Szekeres M, Savinc I, Horvath Z, Saiko P, Pemberger M, Graser G, Bernhaus A,
Ozsvar-Kozma M, Grusch M, Jaeger W, Szekeres T (2008) Biochemical effects of
piceatannol in human HL-60 promyelocytic leukemia cells—synergism with Ara-C. Int J
Oncol 33:887–892
38. Morales P, Haza AI (2012) Selective apoptotic effects of piceatannol and myricetin in human
cancer cells. J Appl Toxicol JAT 32:986–993
39. Suzuki M, Sugimoto K, Tanaka J, Tameda M, Inagaki Y, Kusagawa S, Nojiri K, Beppu T,
Yoneda K, Yamamoto N, Ito M, Yoneda M, Uchida K, Takase K, Shiraki K (2010)
Up-regulation of glypican-3 in human hepatocellular carcinoma. Anticancer Res 30:5055–
5061
40. Klimowicz AC, Bisson SA, Hans K, Long EM, Hansen HC, Robbins SM (2009) The
phytochemical piceatannol induces the loss of CBL and CBL-associated proteins. Mol
Cancer Ther 8:602–614
41. Huang X, Ordemann J, Muller JM, Dubiel W (2012) The COP9 signalosome, cullin 3 and
Keap1 supercomplex regulates CHOP stability and adipogenesis. Biol Open 1:705–710
42. Kwon JY, Seo SG, Heo YS, Yue S, Cheng JX, Lee KW, Kim KH (2012) Piceatannol, natural
polyphenolic stilbene, inhibits adipogenesis via modulation of mitotic clonal expansion and
insulin receptor-dependent insulin signaling in early phase of differentiation. J Biol Chem
287:11566–11578
208 Y.-J. Surh and H.-K. Na
59. Chen WP, Hung LM, Hsueh CH, Lai LP, Su MJ (2009) Piceatannol, a derivative of
resveratrol, moderately slows I(Na) inactivation and exerts antiarrhythmic action in
ischaemia-reperfused rat hearts. Brit J Pharmacol 157:381–391
60. Wang Z, Li J, Cho J, Malik AB (2014) Prevention of vascular inflammation by nanoparticle
targeting of adherent neutrophils. Nat Nanotechnol 9:204–210
61. Yang CJ, Lin CY, Hsieh TC, Olson SC, Wu JM (2011) Control of eotaxin-1 expression and
release by resveratrol and its metabolites in culture human pulmonary artery endothelial cells.
Am J Cardiovasc Dis 1:16–30
62. Kee HJ, Park S, Kang W, Lim KS, Kim JH, Ahn Y, Jeong MH (2014) Piceatannol attenuates
cardiac hypertrophy in an animal model through regulation of the expression and binding of
the transcription factor GATA binding factor 6. FEBS Lett 588:1529–1536
63. Frombaum M, Therond P, Djelidi R, Beaudeux JL, Bonnefont-Rousselot D, Borderie D
(2011) Piceatannol is more effective than resveratrol in restoring endothelial cell
dimethylarginine dimethylaminohydrolase expression and activity after high-glucose
oxidative stress. Free Radic Res 45:293–302
64. Woo A, Min B, Ryoo S (2010) Piceatannol-3’-O-beta-D-glucopyranoside as an active
component of rhubarb activates endothelial nitric oxide synthase through inhibition of
arginase activity. Exp Mol Med 42:524–532
65. Cao L, Li L, Yang H, Yin H (2010) Overexpression of P-selectin glycoprotein ligand-1
enhances adhesive properties of endothelial progenitor cells through Syk activation. Acta
Biochim Biophys Sin 42:507–514
66. Oh KS, Ryu SY, Kim YS, Lee BH (2007) Large conductance Ca2+-activated K+ (BKCa)
channels are involved in the vascular relaxations elicited by piceatannol isolated from Rheum
undulatum rhizome. Planta Med 73:1441–1446
67. Luskova P, Draber P (2004) Modulation of the Fcepsilon receptor I signaling by tyrosine
kinase inhibitors: search for therapeutic targets of inflammatory and allergy diseases. Curr
Pharm Des 10:1727–1737
68. Sato D, Shimizu N, Shimizu Y, Akagi M, Eshita Y, Ozaki S, Nakajima N, Ishihara K,
Masuoka N, Hamada H, Shimoda K, Kubota N (2014) Synthesis of glycosides of resveratrol,
pterostilbene, and piceatannol, and their anti-oxidant, anti-allergic, and neuroprotective
activities. Biosci Biotechnol Biochem 78:1123–1128
69. Ko YJ, Kim HH, Kim EJ, Katakura Y, Lee WS, Kim GS, Ryu CH (2013) Piceatannol
inhibits mast cell-mediated allergic inflammation. Int J Mol Med 31:951–958
70. Matsuda H, Tomohiro N, Hiraba K, Harima S, Ko S, Matsuo K, Yoshikawa M, Kubo M
(2001) Study on anti-Oketsu activity of rhubarb II. Anti-allergic effects of stilbene
components from Rhei undulati Rhizoma (dried rhizome of Rheum undulatum cultivated in
Korea). Biol Pharm Bull 24:264–267
71. Perecko T, Drabikova K, Nosal R, Harmatha J, Jancinova V (2012) Involvement of
caspase-3 in stilbene derivatives induced apoptosis of human neutrophils in vitro. Interdiscip
Toxicol 5:76–80
72. Jancinova V, Perecko T, Nosal R, Svitekova K, Drabikova K (2013) The natural stilbenoid
piceatannol decreases activity and accelerates apoptosis of human neutrophils: involvement
of protein kinase C. Oxid Med Cell Longev 2013:136539
73. Antoine F, Ennaciri J, Girard D (2010) Syk is a novel target of arsenic trioxide (ATO) and is
involved in the toxic effect of ATO in human neutrophils. Toxicol In Vitro 24:936–941
74. Kim DH, Lee YG, Park HJ, Lee JA, Kim HJ, Hwang JK, Choi JM (2015) Piceatannol
inhibits effector T cell functions by suppressing TcR signaling. Int Immunopharmacol
25:285–292
75. Chang JK, Hsu YL, Teng IC, Kuo PL (2006) Piceatannol stimulates osteoblast differentiation
that may be mediated by increased bone morphogenetic protein-2 production. Eur J
Pharmacol 551:1–9
76. Ke K, Sul OJ, Rajasekaran M, Choi HS (2015) MicroRNA-183 increases osteoclastogenesis
by repressing heme oxygenase-1. Bone 81:237–246
210 Y.-J. Surh and H.-K. Na
95. Roupe KA, Yanez JA, Teng XW, Davies NM (2006) Pharmacokinetics of selected stilbenes:
rhapontigenin, piceatannol and pinosylvin in rats. J Pharm Pharmacol 58:1443–1450
96. Miksits M, Maier-Salamon A, Vo TP, Sulyok M, Schuhmacher R, Szekeres T, Jager W
(2010) Glucuronidation of piceatannol by human liver microsomes: major role of UGT1A1,
UGT1A8 and UGT1A10. J Pharm Pharmacol 62:47–54
97. Miksits M, Sulyok M, Schuhmacher R, Szekeres T, Jager W (2009) In-vitro sulfation of
piceatannol by human liver cytosol and recombinant sulfotransferases. J Pharm Pharmacol
61:185–191
98. Chun YJ, Kim MY, Guengerich FP (1999) Resveratrol is a selective human cytochrome
P450 1A1 inhibitor. Biochem Biophys Res Commun 262:20–24
99. Chen ZH, Hurh YJ, Na HK, Kim JH, Chun YJ, Kim DH, Kang KS, Cho MH, Surh YJ
(2004) Resveratrol inhibits TCDD-induced expression of CYP1A1 and CYP1B1 and
catechol estrogen-mediated oxidative DNA damage in cultured human mammary epithelial
cells. Carcinogenesis 25:2005–2013
100. Macpherson L, Matthews J (2010) Inhibition of aryl hydrocarbon receptor-dependent
transcription by resveratrol or kaempferol is independent of estrogen receptor alpha
expression in human breast cancer cells. Cancer Lett 299:119–129
101. Chang TK, Chen J, Yu CT (2007) In vitro inhibition of rat CYP1A1 and CYP1A2 by
piceatannol, a hydroxylated metabolite of trans-resveratrol. Drug Metab Lett 1:13–16
102. Mikstacka R, Rimando AM, Szalaty K, Stasik K, Baer-Dubowska W (2006) Effect of natural
analogues of trans-resveratrol on cytochromes P4501A2 and 2E1 catalytic activities.
Xenobiotica 36:269–285
103. Oskarsson A, Spatafora C, Tringali C, Andersson AO (2014) Inhibition of CYP17A1 activity
by resveratrol, piceatannol, and synthetic resveratrol analogs. Prostate 74:839–851
104. Messiad H, Amira-Guebailia H, Houache O (2013) Reversed phase high performance liquid
chromatography used for the physicochemical and thermodynamic characterization of
piceatannol/beta-cyclodextrin complex. J Chromatogr B Analyt Technol Biomed Life Sci
926:21–27
Chapter 10
Fisetin and Its Role in Chronic Diseases
Keywords Fisetin Inflammation Chronic diseases Cytokines Transcription
factors Nutraceuticals Neurological diseases
Cardiovascular diseases
Pulmonary diseases Diabetes Obesity Allergy Cancer
Abbreviations
AChE Acetylcholinesterase
CAT Catalase
COX Cyclooxygenase
EMT Epithelial-to-mesenchymal transition
EPCR Endothelial cell protein C receptor
ERK Extracellular signal-regulated kinase
GABAA Gamma-aminobutyric acid A
GLUT4 Glucose transporter type 4
GR Glutathione reductase
GSH Glutathione
GSH-Px Glutathione peroxidase
GST Glutathione S-transferase
HDL High density lipoprotein
HMGB1 High mobility group box 1
IL Interleukin
iNOS Inducible nitrogen oxide synthase
LDL Low-density lipoprotein
LOX Lipoxygenase
MAPKs Mitogen-activated protein kinases
MMP Matrix metalloproteinase
MPO Myeloperoxidase
mTOR Mammalian target of rapamycin
NFjB Nuclear factor-kappa B
NO Nitric oxide
Nrf2 Nuclear factor erythroid-2-related factor 2
PECAM-1 Platelet endothelial cell adhesion molecule 1
PGE2 Prostaglandin E2
PI3K Phosphatidylinositol 3-kinase
PPARc Peroxisome proliferator-activated receptor gamma
SCD-1 Stearoyl-CoA desaturase-1
SCEM Small Clot Embolism Model
SOD Superoxide dismutase
SREBP1C Sterol-regulatory-element-binding protein-1c
STAT Signal transducer and activator of transcription
TARC Thymus and activation regulated chemokine
TLR4 Toll-like receptor 4
TSLP Thymic stromal lymphopoietin
VLDL Very-low-density lipoprotein
10 Fisetin and Its Role in Chronic Diseases 215
10.1 Introduction
found in the Quebracho colorado and Rhus cotinus, lac tree (Rhus vemiciflua
Stokes) extract, smoke tree (Cotinus coggygria), Pinopyta species like Callitropsis
nootkatensis (yellow cypress) and other trees and shrubs (Fig. 10.3) [15]. Fisetin is
a potent antioxidant and free radical scavenger. It has shown potential to inhibit cell
proliferation, growth, and survival of various cancer cells via different mechanisms
[16–21]. Its anti-invasive and anti-angiogenic effects were also recently reported
[22–24]. A growing body of evidence has demonstrated that fisetin has potential to
prevent and/or inhibit various chronic inflammation-related conditions [25–30].
Neuroprotective, cardioprotective, and anti-diabetic potentials of fisetin have been
established by employing cell culture studies and animal models relevant to human
diseases [26, 31–37]. More importantly, treatment of these animals with fisetin was
devoid of any sign of measurable toxicity. Using experimental animals, studies
have demonstrated that fisetin was readily absorbed and distributed to the blood
vessels [38]. Moreover, studies have demonstrated that after 40 min of oral
administration, fisetin can be detected within the blood vessels of the brain for 2 h
suggesting that it is well absorbed and bioavailable in the distal organs [38]. Due to
low toxicity and a wide range of beneficial pharmacological effects, fisetin has been
accepted as a nutraceutical and nutritional dietary supplement for neuroprotection.
As the medical sciences progress, we are beginning to understand with greater
detail the mechanisms by which inflammation, cancer, and chronic disease pro-
gress. Despite having our greater understanding, many chronic disease processes
continue to evade and defy modern therapies. For this reason, novel approaches to
the management of chronic disease are necessary. Fisetin is a natural compound that
218 H.C. Pal et al.
has evoked a great amount of research interest in recent years, demonstrating potent
anti-inflammatory, anti-tumorigenic, and anti-oxidant properties. In this chapter, we
discuss various pharmacological effects of fisetin against different pathological
conditions with special emphasis on diseases related to chronic inflammatory
conditions (Table 10.1).
Studies have demonstrated that fisetin inhibits proliferation of various cancer cells
in vitro and in vivo. Fisetin at lower doses targets Aurora B kinase by inhibiting
kinetochore and centromere localization leading to immature segregation of chro-
mosomes and premature cessation of mitosis without cytokinesis resulting in
10 Fisetin and Its Role in Chronic Diseases 221
potential to protect the brain due to their ability to modulate intracellular signals
promoting cellular survival [49–51]. Fisetin effectively protected central nervous
system-derived nerve cells (HT-22 cells) and rat primary neurons from glutamate
toxicity, hypoglycemia, and oxidative injuries by altering glutathione
(GSH) metabolism. In addition, fisetin blocked hydrogen peroxide (H2O2)-induced
neuronal death by reactive oxygen species (ROS) scavenging effects [52]. Fisetin
also inhibited in vitro myelin phagocytosis by macrophages responsible for
secretion of inflammatory mediators, suggesting the potential to reduce the risk of a
chronic inflammatory disease of central nervous system called multiple sclerosis
leading to neurological deficits [53]. In addition, fisetin treatment significantly
reduced ROS production without affecting the viability of macrophage cells. Upon
in vitro experimental evaluation of a variety of flavonoids using a well-studied
model system of neuronal differentiation PC-12 cells expressing nerve growth
factor (NGF) derived from rat embryonic origin from the neural crest, Sagara et al.
[54] found that fisetin was the most effective neuroprotective flavonoid. Employing
an extensive mechanistic approach, it was found that fisetin most effectively
induced neurite outgrowth and PC-12 cell differentiation by activation of the
Ras-ERK cascade, particularly by ERK activation [32, 54].
It was also shown that fisetin promotes nerve cell survival by enhancement of
proteasome activity after neurotrophic factor withdrawal, suggesting that it can also
act as neurotrophic factor [55]. Most importantly, Maher et al. [34] found that
fisetin treatment enhances memory in experimental rats by activation of ERK and
induction of cAMP response element-binding protein (CREB) phosphorylation in
rats hippocampal slices. Moreover, intraperitoneal administration of a single dose of
liposomal preparation containing 30 mg/kg dose of fisetin resulted in detection of
8.23 ng fisetin per gram of brain tissue and exerted protective effects demonstrated
by recovery of the cytoarchitecture in ischemic areas of striatum and cortex in rats.
However, a similar dose of fisetin went undetected in the brain when administered
in aqueous preparation [56]. Similarly, intravenous injection of 50 mg/kg fisetin
initiated after 5 min of embolizaion resulted in significant protection in Rabbit
Small Clot Embolism Model (SCEM) [55]. Moreover, oral administration of 5–
25 mg/kg of fisetin resulted in dose-dependent enhancement in long-term memory
in mice [34]. Furthermore, feeding of mice for 10 months with diet containing
fisetin (500 mg/kg of food) resulted in substantial improvement in learning com-
pared to age-matched mice fed on a fisetin-free diet. Moreover, feeding of
fisetin-containing diet to these mice resulted in significantly improved memory in
the morris water maze (MWM) test relative to age-matched mice fed on a
fisetin-free diet [48]. In addition, studies have demonstrated that feeding of 14.8 gm
of dried aqueous strawberry extract (a major source of fisetin) per kilogram of diet
for 8 weeks to 19-months old rats indicated that fisetin-containing strawberry
extract reversed age-related deficits and improved memory in the MWM compared
to control diet fed rats [57]. Feeding of strawberry extract containing diet to rats
demonstrated better protection against spatial deficits caused by irradiation with
1.5 Gy of 1 GeV/n 56Fe particles. The results from this study provided evidence
that strawberry extract fed animals were better able to retain place information (a
10 Fisetin and Its Role in Chronic Diseases 223
In the United States, obesity is one of the greatest public health concerns of the
twenty-first century. Several mechanisms have been suggested by which fisetin
treatment may mitigate the pathogenesis of diet-induced obesity. Preliminary evi-
dence suggests that fisetin supplementation may reduce the risk of developing
obesity by decreasing differentiation and proliferation of adipocytes.
Undifferentiated fibroblasts, or preadipocytes, have been identified as a potential
target of fisetin treatment. Recent studies have demonstrated that differentiation of
3T3-L1 undifferentiated fibroblasts into adipocytes is reduced by fisetin [75, 76].
One study found that fisetin treatment reduced phosphorylation of mTORC1 and
upstream promoters of mTORC1 signaling including AKT and S6K1. In vivo
experiments confirmed these findings; in murine models fed high-fat diets; fisetin
supplementation reduced weight gains and accumulation of white adipose tissue via
suppression of mTORC1 signaling and reduced differentiation of preadipocytes
[75]. Another study suggested that fisetin treatment decreased adipocyte differen-
tiation and proliferation by suppressing mitotic clonal expansion. Fisetin treatment
reduced expression of several key cell cycle promoters including cyclin A, cyclin
D1, and cdk4. In addition, fisetin upregulated the cell cycle inhibitor p27, pro-
moting a sustained G0 phase [76].
Fisetin may play a role in obesity treatment or prevention by modulating
cholesterol homeostasis. In Sprague-Dawley hypercholesterolemic rat models,
treatment with fisetin reduced several critical markers of obesity risk. The blood
lipid profile of rats on a high-fat diet was improved by fisetin treatment; total
cholesterol, LDL, HDL, and hepatic cholesterol levels were all reduced. The
hepatic abundance of CYP7A1 was reduced to near control levels after fisetin
treatment, suggesting a return to normal bile acid metabolism. Another possible
mechanism by which fisetin may regulate obesity pathogenesis is by reducing
hepatic lipogenesis. In Sprague-Dawley rats fed with high-fat diets, fisetin reduced
expression of hepatic mRNA associated with lipogenesis including PPARc,
SREBP1C, and SCD-1 compared to controls. Expression of gene products asso-
ciated with fatty acid synthesis including fatty acid synthase and ATP citrate lyase
were also markedly reduced by fisetin treatment. In addition, fisetin-induced
expression of GLUT4 in 3T3-L1 differentiated adipose cells, decreasing concen-
trations of serum glucose [77, 78]. Another study found that fisetin treatment
inhibited high-fat diet-induced expression of miR-378 and PGC-1B resulting in
decreased hepatic fat accumulation and reversal of metabolic enzyme dysregula-
tion [79]. These data indicate that fisetin treatment may attenuate obesity by
reducing hepatic lipid biosynthesis.
10 Fisetin and Its Role in Chronic Diseases 229
Basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs) are the most
frequently diagnosed non-melanoma skin cancers (NMSCs). Ultraviolet
(UV) irradiation is considered the most important extrinsic factor contributing to
inflammation and skin cancer. Consumption of nontoxic dietary flavonoids to
potentially prevent skin cancers has drawn a great deal of attention. Treatment of
human epidermoid carcinoma A431 cells with fisetin resulted in decease in cell
proliferation. Fisetin treatment enhanced G2/M cell population and induced apop-
tosis through disruption of mitochondrial membrane potential and modulation in
Bcl-2 family proteins. Fisetin treatment also promoted release of cytochrome c and
230 H.C. Pal et al.
Prostate cancer is one of the most common cancers and leading causes of death in
men [95, 102]. Consumption of flavonoid-rich diets in East Asian countries such as
China and Japan has been associated with a 60- to 80-fold lower incidence and
reduced mortality of prostate cancer [103]. Fisetin has been found to be effective
against prostate cancer. In vitro studies using fisetin have demonstrated that fisetin
inhibits cell proliferation and induces cell cycle arrest and apoptosis in
androgen-sensitive human prostate LNCaP and CWR22Rʋ1 cells as well as in
androgen receptor (AR)-negative prostate cancer PC-3 cells [103, 104].
Furthermore, fisetin treatment inhibited viability and colony formation of a
P-glycoprotein-overexpressing multi-drug resistant cancer cell line NCI/ADR-RE
[105]. Importantly, fisetin exhibited minimal cytotoxic effects on normal prostate
epithelial cells (PrECs). Fisetin treatment induced cell cycle arrest at G2/M phase in
PC-3 cells; whereas LNCaP cells were arrested at G1 phase of cell cycle. G1-phase
cell cycle arrest in LNCaP cells induced by fisetin treatment was associated with
reduced protein expression of cyclins D1, D2, and E. Fisetin treatment also reduced
the protein expression of cdks 2, 4, and 6 with simultaneous increase in protein
expression of WAF1/p21 and KIP1/p27. Furthermore, fisetin treatment induced
apoptosis in LNCaP cells through induction of pro-apoptotic proteins (Bax, Bak,
Bad, and Bid) and inhibition of anti-apoptotic proteins (Bcl-2 and Bcl-xL), cyto-
chrome c release, activation of caspases (3, 8, and 9) and cleavage of PARP. In
addition, fisetin treatment also reduced protein expression of upstream regulators of
apoptosis such as PI3 K and decreased the phosphorylation of AKT at Ser473 and
Thr308, which are involved in cell proliferation and survival [104]. Fisetin induced
PC3 cell death by induction of autophagy, as observed by an increase in LC3 II
protein expression. Fisetin treatment of PC-3 cells resulted in inhibition of mTOR
kinase activity, basal expression of mTOR and autophosphorylation of mTOR at
Ser2481. It also inhibited formation of mTORC1/2 complexes via downregulation of
Raptor, Rictor, PRAS40, and GbL protein expression. Fisetin treatment also
inhibited the activation of p70-S6 kinase (S6K70) and increased expression of
eukaryotic translation initiation factor 4E-binding protein 1(4EBP1) by its
dephosphorylation from hyperphosphorylated c form to the hypo- or
non-phosphorylated a form. Furthermore, fisetin treatment of PC-3 cells disrupted
assembly of translation complex by increasing eIF4E bound 4EBP1 and simulta-
neous reduction in eIF4G binding to eIF4E [20].
Studies have demonstrated that combination of fisetin with TRAIL resulted in
enhanced apoptosis of TRAIL-resistant androgen-dependent LNCaP cells and the
androgen-independent DU145 and PC-3 cells [44]. In TRAIL-resistant LNCaP
cells, fisetin treatment increased the expression of TRAIL-R1 and reduced NFjB
activity. Moreover, studies have demonstrated that fisetin (5–20 lM) inhibited
adhesion, migration, and invasion of highly metastatic PC-3 cells [46, 105]. Fisetin
treatment significantly reduced protein expression as well as mRNA expression of
MMP-2 and MMP-9 involved in the degradation of ECM to facilitate invasion and
10 Fisetin and Its Role in Chronic Diseases 233
Fisetin treatment also inhibited activation and translocation of NFjB, which are
required for stimulation of COX-2 expression. PGE2 secretion was also reduced as
a consequence of COX-2 inhibition by fisetin in HT-29 cells. Fisetin did not affect
EP-2 and EP-4 expression, suggesting that fisetin inhibits COX-2/PGE2 signaling
through regulating ligand availability. Moreover, fisetin treatment also inhibited
phosphorylation of EGFR at the Tyr1068 residue in HT-29 cells as a consequence of
PGE2 inhibition, which is a known transactivator of EGFR leading to promotion of
tumor growth and invasion [109]. Furthermore, depletion of Securin expression
(also known as pituitary tumor transforming gene and acts as a marker of inva-
siveness in colon cancers) sensitized human colon cancer cells to fisetin-induced
apoptosis. Phosphorylation of p53 and cleavage of caspase-3 and PARP were
enhanced in HCT116 securin-null cells or in wild-type cells in which securin was
knock down [110]. Studies have also demonstrated that the apoptotic effect of
fisetin on colon cancer cells (COLO205, HCT-116, HT-29, and HCT-15) is
enhanced with N-Acetyl-L-Cysteine treatment, suggesting that fisetin-induced
apoptosis in colon cancer cells is independent of ROS induction [111, 112].
Lung cancer is the most deadly cancer in the United States and worldwide. Tobacco
smoking is considered as the most important risk factor for lung cancer develop-
ment. Results from clinical and epidemiologic studies have demonstrated a strong
association between chronic inflammation and lung cancer [113, 114]. A growing
body of evidence has demonstrated that tobacco smoke exposure induces car-
cinogenic inflammatory responses and mutagenic effects in the lungs. Infiltration of
inflammatory cells in the lungs is the initial pathological hallmark of smoking.
These inflammatory macrophages and neutrophils produce pro-inflammatory
mediators and cytokines to further enhance the inflammatory condition and pro-
mote tumor growth [115, 116]. Fisetin treatment significantly inhibited lung cancer
cell proliferation but had minimal toxic effects on normal human embryonic
epithelial cells at physiologically achievable concentration [16, 117]. Fisetin
treatment induced intracellular ROS production, mitochondrial membrane depo-
larization, and apoptosis in NSCLC cells with an increase in Sub-G1 cell popula-
tion. Fisetin treatment resulted in reduced expression of Bcl-2 and enhanced
expression of Bax, as well as activation of caspase-3 and -9 [118]. Relatively
nontoxic concentrations of fisetin inhibited adhesion, invasion, and migration of
A549 cells. Gelatin and casein zymography experiments demonstrated that fisetin
inhibited MMP-2 and u-PA at protein and mRNA levels. In addition, fisetin
decreased ERK1/2 phosphorylation, however it did not affect the phosphorylation
of JNK1/2 and p38. Moreover, fisetin inhibited DNA binding activities of tran-
scription factors NFjB and AP-1 (c-Fos, and c-Jun) in A549 cells [117].
Furthermore, fisetin treatment to lung cancer cells inhibited the expression of
regulatory (p85) and catalytic (p110) subunits of PI3 K and phosphorylation of
10 Fisetin and Its Role in Chronic Diseases 235
AKT both at Ser473 and Thr308 moieties. Fisetin treatment also activated PTEN, a
negative regulator of PI3 K signaling, and increased phosphorylation of AMPKa
kinase thus inhibiting protein translation by mTOR. Fisetin treatment also inhibited
the phosphorylation of mTOR at Ser2448 as a consequence of inhibition of AKT
phosphorylation [16].
In experimental lung carcinogenesis, fisetin inhibited benzo(a)pyrene-induced
lung cancer development in Swiss albino mice [119]. Histological evaluation of
lungs revealed that fisetin treatment significantly reduced the degree of histological
lesions with reduced cell proliferation. Biochemical analysis demonstrated that
fisetin treatment restored enzymatic and nonenzymatic antioxidants. Furthermore,
evaluation of mitochondrial specific enzymes and tumor markers demonstrated that
fisetin treatment inhibited production of isocitrate dehydrogenase, a-ketoglutarate
dehydrogenase, succinate dehydrogenase, malate dehydrogenase and carcinogenic
embryonic tumor antigen in benzo(a)pyrene-induced lung carcinogenesis [120]. In
addition, fisetin treatment resulted in release of cytochrome c and activation of
caspase-3. Furthermore, fisetin treatment inhibited viability of Lewis lung carci-
noma (LLC) and endothelial cells (EAhy 926), with minimum effect on normal NIH
3T3 cells. NIH3T3 cells were five times less sensitive to fisetin than either LLC or
endothelial cells, demonstrating that fisetin specifically targets cancer cells and
endothelial cells involved in tumor angiogenesis [121]. Fisetin treatment to LLC
cells induced apoptosis and accumulation of cells in G2/M phase with concomitant
decrease in G1 phase. Endothelial cells (EAhy 926) were more sensitive to fisetin
treatment with increase in sub-G1cells and decrease in G1, S, and G2/M cells.
Fisetin treatment also inhibited migration and capillary-like structure-forming
abilities of endothelial cells and inhibited tumor growth and angiogenesis in vivo as
demonstrated by reduced expression of PECAM-1. Moreover, antitumor activity of
fisetin was enhanced in combination with cyclophosphamide [121].
Intraperitoneal administration of 1 or 3 mg/kg fisetin in BALB/c mice inhibited
ovalbumin-induced allergic asthma, which is a chronic disease of lung inflamma-
tion, airway hyper-responsiveness and mucus overproduction associated with the
bronchial epithelium, mucus-secreting glands and lung parenchyma [122, 123].
Treatment of fisetin in experimental asthma mouse model resulted in inhibition of
lung inflammation, goblet cell hyperplasia, and airway hyper-responsiveness. These
effects were associated with a decrease in eosinophils and lymphocytes in bron-
choalveolar lavage fluid. In addition, fisetin treatment reduced expression of
eotaxin-1 and thymic stromal lymphopoietin (TSLP) (key initiators of allergic
airway inflammation), IL-4, IL-5, and IL-13 (Th2-associated cytokines) production
in lungs. Fisetin treatment also inhibited mRNA expression of adhesion molecules,
chitinase, IL-17, IL-33, Muc5ac, and iNOS, as wells as eosinophilia and airway
mucus production in lung tissue induced by ovalbumin. In addition, fisetin treat-
ment also inhibited expression of Th2-predominant transcription factor GATA-3
and cytokines in thoracic lymph node cells and splenocytes. Moreover, in
TNFa-stimulated bronchial epithelial cells and OVA-stimulated lung tissues, fisetin
inhibited activation of NFjB by blocking nuclear translocation of subunit p65 and
DNA binding activity [122, 123]. A recent study demonstrated that fisetin inhibits
236 H.C. Pal et al.
and tissue remodeling by excessive collagen accumulation due to IFNc and IL-2
(Th1-dominat immune response) is associated with delayed-type hyper-sensitivity.
A recent study by Kim et al. [25] demonstrated that oral administration of fisetin at
20 or 50 mg/kg daily from days 8 to 15, significantly inhibited DNFB-induced
atopic dermatitis-like clinical symptoms such as erythema, edema, oozing, and
excoriation in NC/Nga mice. Fisetin treatment also inhibited DNFB-induced epi-
dermal thickness and infiltration of eosinophils, mast cells, CD4+ T and CD8+ T
cells in ear and dorsal skin. Moreover, fisetin treatment also reduced expression of
Th2 cytokines IL-5, IL-13, TNFa, thymus, and activation regulated chemokine
(TARC) and TSLP mRNA expression produced by dermal leukocytes and ker-
atinocytes. Furthermore, fisetin treatment suppressed production of IFNc and IL-4
by the activated lymph node CD4+ T cells with increased production of IL-10. In
addition, fisetin inhibited activation of NFjB by reducing levels of phosphorylated
p65 [25].
Studies on HUVECs and septic mice have demonstrated that fisetin inhibited
sepsis-related mortality. Fisetin treatment inhibited hyperpermeability and leuko-
cyte migration in septic mice induced by LPS and cecal ligation and puncture
(CLP)-mediated release of high mobility group box 1 (HMGB1) protein. In addi-
tion, fisetin treatment greatly inhibited PMA and CLP-induced expression of
endothelial cell protein C receptor involved in vascular inflammation. Furthermore,
fisetin treatment also inhibited production of TNFa and IL-1b as well as activation
of AKT, NFjB, and ERK1/2 in HUVEC cells induced by HMGB1 [30]. In
addition, in vitro and in vivo studies have demonstrated that fisetin inhibited high
glucose-induced vascular inflammation, vascular permeability, leukocyte adhesion,
and migration, cell adhesion molecule expression, ROS formation and NFjB
activation [26].
A recent study by Kim et al. [130] demonstrated that fisetin suppresses
macrophage-mediated inflammation by blocking Src and Syk, the major NFjB
regulatory protein tyrosine kinases. Fisetin treatment of RAW264.7 cells inhibited
LPS-induced production of NO, transcriptional activation of inflammatory genes
(iNOS, COX-2, and TNFa) and activation of NFjB without any cytotoxic effect on
these cells. Moreover, the autophosphorylation levels of Src and Syk were signif-
icantly suppressed without decreasing total levels of Src and Syk [130].
10.5 Conclusions
Acknowledgments The work highlighted from the author’s laboratory was supported by NIH
Grant R21CA173043.
References
16. Khan N, Afaq F, Khusro FH, Mustafa Adhami V, Suh Y, Mukhtar H (2012) Dual inhibition
of phosphatidylinositol 3-kinase/Akt and mammalian target of rapamycin signaling in human
nonsmall cell lung cancer cells by a dietary flavonoid fisetin. Int J Cancer 130(7):1695–1705
17. Khan N, Afaq F, Mukhtar H (2008) Cancer chemoprevention through dietary antioxidants:
progress and promise. Antioxid Redox Signal 10(3):475–510
18. Pal HC, Athar M, Elmets CA, Afaq F (2015) Fisetin inhibits UVB-induced cutaneous
inflammation and activation of PI3 K/AKT/NFjB signaling pathways in SKH-1 hairless
mice. Photochem Photobiol 91(1):225–234
19. Pal HC, Sharma S, Elmets CA, Athar M, Afaq F (2013) Fisetin inhibits growth, induces G2/
M arrest and apoptosis of human epidermoid carcinoma A431 cells: role of mitochondrial
membrane potential disruption and consequent caspases activation. Exp Dermatol 22
(7):470–475
20. Suh Y, Afaq F, Khan N, Johnson JJ, Khusro FH, Mukhtar H (2010) Fisetin induces
autophagic cell death through suppression of mTOR signaling pathway in prostate cancer
cells. Carcinogenesis 31(8):1424–1433
21. Syed DN, Afaq F, Maddodi N, Johnson JJ, Sarfaraz S, Ahmad A, Setaluri V, Mukhtar H
(2011) Inhibition of human melanoma cell growth by the dietary flavonoid fisetin is
associated with disruption of Wnt/b-catenin signaling and decreased Mitf levels. J Invest
Dermatol 131(6):1291–1299
22. Pal HC, Baxter RD, Hunt KM, Agarwal J, Elmets CA, Athar M, Afaq F (2015) Fisetin, a
phytochemical, potentiates sorafenib-induced apoptosis and abrogates tumor growth in
athymic nude mice implanted with BRAF-mutated melanoma cells. Oncotarget. 6
(29):28296–28311
23. Pal HC, Diamond AC, Strickland LR, Kappes JC, Katiyar SK, Elmets CA, Athar M, Afaq F
(2016) Fisetin, a dietary flavonoid, augments the anti-invasive and anti-metastatic potential of
sorafenib in melanoma. Oncotarget. 7(2):1227–1241.
24. Pal HC, Sharma S, Strickland LR, Katiyar SK, Ballestas ME, Athar M, Elmets CA, Afaq F
(2014) Fisetin inhibits human melanoma cell invasion through promotion of mesenchymal to
epithelial transition and by targeting MAPK and NFjB signaling pathways. PLoS ONE 9(1):
e86338
25. Kim GD, Lee SE, Park YS, Shin DH, Park GG, Park CS (2014) Immunosuppressive effects
of fisetin against dinitrofluorobenzene-induced atopic dermatitis-like symptoms in NC/Nga
mice. Food Chem Toxicol 66:341–349
26. Kwak S, Ku SK, Bae JS (2014) Fisetin inhibits high-glucose-induced vascular inflammation
in vitro and in vivo. Inflamm Res 63(9):779–787
27. Lee JD, Huh JE, Jeon G, Yang HR, Woo HS, Choi DY, Park DS (2009) Flavonol-rich
RVHxR from Rhus verniciflua Stokes and its major compound fisetin inhibits
inflammation-related cytokines and angiogenic factor in rheumatoid arthritic fibroblast-like
synovial cells and in vivo models. Int Immunopharmacol 9(3):268–276
28. Park DK, Lee YG, Park HJ (2013) Extract of Rhus verniciflua bark suppresses
2,4-dinitrofluorobenzene-induced allergic contact dermatitis. Evid Based Complement
Alternat 2013:879696
29. Park HH, Lee S, Oh JM, Lee MS, Yoon KH, Park BH, Kim JW, Song H, Kim SH (2007)
Anti-inflammatory activity of fisetin in human mast cells (HMC-1). Pharmacol Res 55
(1):31–37
30. Yoo H, Ku SK, Han MS, Kim KM, Bae JS (2014) Anti-septic effects of fisetin in vitro and
in vivo. Inflammation. 37(5):1560–1574
31. Currais A, Prior M, Dargusch R, Armando A, Ehren J, Schubert D, Quehenberger O,
Maher P (2014) Modulation of p25 and inflammatory pathways by fisetin maintains
cognitive function in Alzheimer’s disease transgenic mice. Aging Cell 13(2):379–390
32. Maher P (2006) A comparison of the neurotrophic activities of the flavonoid fisetin and some
of its derivatives. Free Radic Res 40(10):1105–1111
33. Maher P (2008) The flavonoid fisetin promotes nerve cell survival from trophic factor
withdrawal by enhancement of proteasome activity. Arch Biochem Biophys 476(2):139–144
240 H.C. Pal et al.
34. Maher P, Akaishi T, Abe K (2006) Flavonoid fisetin promotes ERK-dependent long-term
potentiation and enhances memory. Proc Natl Acad Sci USA 103(44):16568–16573
35. Maher P, Dargusch R, Bodai L, Gerard PE, Purcell JM, Marsh JL (2011) ERK activation by
the polyphenols fisetin and resveratrol provides neuroprotection in multiple models of
Huntington’s disease. Hum Mol Genet 20(2):261–270
36. Maher P, Dargusch R, Ehren JL, Okada S, Sharma K, Schubert D (2011) Fisetin lowers
methylglyoxal dependent protein glycation and limits the complications of diabetes.
PLoS ONE 6(6):e21226
37. Renoudet VV, Costa-Mallen P, Hopkins E (2012) A diet low in animal fat and rich in
N-hexacosanol and fisetin is effective in reducing symptoms of Parkinson’s disease. J Med
Food 15(8):758–761
38. Krasieva TB, Ehren J, O’Sullivan T, Tromberg BJ, Maher P (2015) Cell and brain tissue
imaging of the flavonoid fisetin using label-free two-photon microscopy. Neurochem Int
89:243–248
39. Gollapudi P, Hasegawa LS, Eastmond DA (2014) A comparative study of the aneugenic and
polyploidy-inducing effects of fisetin and two model Aurora kinase inhibitors. Mutat Res,
Genet Toxicol Environ Mutagen 767:37–43
40. Lopez-Lazaro M, Willmore E, Austin CA (2010) The dietary flavonoids myricetin and fisetin
act as dual inhibitors of DNA topoisomerases I and II in cells. Mutat Res 696(1):41–47
41. Olaharski AJ, Mondrala ST, Eastmond DA (2005) Chromosomal malsegregation and
micronucleus induction in vitro by the DNA topoisomerase II inhibitor fisetin. Mutat Res 582
(1–2):79–86
42. Salmela AL, Pouwels J, Varis A, Kukkonen AM, Toivonen P, Halonen PK, Perälä M,
Kallioniemi O, Gorbsky GJ, Kallio MJ (2009) Dietary flavonoid fisetin induces a forced exit
from mitosis by targeting the mitotic spindle checkpoint. Carcinogenesis 30(6):1032–1040
43. Sung B, Pandey MK, Aggarwal BB (2007) Fisetin, an inhibitor of cyclin-dependent kinase 6,
down-regulates nuclear factor-kappaB-regulated cell proliferation, antiapoptotic and
metastatic gene products through the suppression of TAK-1 and receptor-interacting
protein-regulated IkappaBalpha kinase activation. Mol Pharmacol 71(6):1703–1714
44. Szliszka E, Helewski KJ, Mizgala E, Krol W (2011) The dietary flavonol fisetin enhances the
apoptosis-inducing potential of TRAIL in prostate cancer cells. Int J Oncol 39(4):771–779
45. Khan N, Asim M, Afaq F, Abu Zaid M, Mukhtar H (2008) A novel dietary flavonoid fisetin
inhibits androgen receptor signaling and tumor growth in athymic nude mice. Cancer Res 68
(20):8555–8563
46. Chien CS, Shen KH, Huang JS, Ko SC, Shih YW (2010) Antimetastatic potential of fisetin
involves inactivation of the PI3 K/Akt and JNK signaling pathways with downregulation of
MMP-2/9 expressions in prostate cancer PC-3 cells. Mol Cell Biochem 333(1–2):169–180
47. Chuang JY, Chang PC, Shen YC, Lin C, Tsai CF, Chen JH, Yeh WL, Wu LH, Lin HY,
Liu YS, Lu DY (2014) Regulatory effects of fisetin on microglial activation. Molecules 19
(7):8820–8839
48. Maher P (2009) Modulation of multiple pathways involved in the maintenance of neuronal
function during aging by fisetin. Genes Nutr 4(4):297–307
49. Dajas F, Rivera F, Blasina F, Arredondo F, Echeverry C, Lafon L, Morquio A, Heinzen H
(2003) Cell culture protection and in vivo neuroprotective capacity of flavonoids. Neurotox
Res 5(6):425–432
50. Dajas F, Rivera-Megret F, Blasina F, Arredondo F, Abin-Carriquiry JA, Costa G,
Echeverry C, Lafon L, Heizen H, Ferreira M, Morquio A (2003) Neuroprotection by
flavonoids. Braz J Med Biol Res 36(12):1613–1620
51. Echeverry C, Arredondo F, Martínez M, Abin-Carriquiry JA, Midiwo J, Dajas F (2015)
Antioxidant activity, cellular bioavailability, and iron and calcium management of
neuroprotective and nonneuroprotective flavones. Neurotox Res 27(1):31–42
52. Ishige K, Schubert D, Sagara Y (2001) Flavonoids protect neuronal cells from oxidative
stress by three distinct mechanisms. Free Radic Biol Med 30(4):433–446
10 Fisetin and Its Role in Chronic Diseases 241
53. Hendriks JJ, de Vries HE, van der Pol SM, van den Berg TK, van Tol EA, Dijkstra CD
(2003) Flavonoids inhibit myelin phagocytosis by macrophages; a structure–activity
relationship study. Biochem Pharmacol 65(5):877–885
54. Sagara Y, Vanhnasy J, Maher P (2004) Induction of PC12 cell differentiation by flavonoids is
dependent upon extracellular signal-regulated kinase activation. J Neurochem 90
(5):1144–1155
55. Maher P, Salgado KF, Zivin JA, Lapchak PA (2007) A novel approach to screening for new
neuroprotective compounds for the treatment of stroke. Brain Res 1173:117–125
56. Rivera F, Urbanavicius J, Gervaz E, Morquio A, Dajas F (2004) Some aspects of the in vivo
neuroprotective capacity of flavonoids: bioavailability and structure-activity
relationship. Neurotox Res 6(7–8):543–553
57. Joseph JA, Shukitt-Hale B, Denisova NA, Bielinski D, Martin A, McEwen JJ, Bickford PC
(1999) Reversals of age-related declines in neuronal signal transduction, cognitive, and
motor behavioral deficits with blueberry, spinach, or strawberry dietary supplementation.
J Neurosci 19(18):8114–8121
58. Shukitt-Hale B, Carey AN, Jenkins D, Rabin BM, Joseph JA (2007) Beneficial effects of fruit
extracts on neuronal function and behavior in a rodent model of accelerated aging. Neurobiol
Aging 28(8):1187–1194
59. Zheng LT, Ock J, Kwon BM, Suk K (2008) Suppressive effects of flavonoid fisetin on
lipopolysaccharide-induced microglial activation and neurotoxicity. Int Immunopharmacol 8
(3):484–494
60. Tahanian E, Sanchez LA, Shiao TC, Roy R, Annabi B (2011) Flavonoids targeting of IjB
phosphorylation abrogates carcinogen-induced MMP-9 and COX-2 expression in human
brain endothelial cells. Drug Des Devel Ther 5:299–309
61. Zhen L, Zhu J, Zhao X, Huang W, An Y, Li S, Du X, Lin M, Wang Q, Xu Y, Pan J (2012)
The antidepressant-like effect of fisetin involves the serotonergic and noradrenergic system.
Behav Brain Res 228(2):359–366
62. Inkielewicz-Stepniak I, Radomski MW, Wozniak M (2012) Fisetin prevents fluoride- and
dexamethasone-induced oxidative damage in osteoblast and hippocampal cells. Food Chem
Toxicol 50(3–4):583–589
63. Prakash D, Gopinath K, Sudhandiran G (2013) Fisetin enhances behavioral performances
and attenuates reactive gliosis and inflammation during aluminum chloride-induced
neurotoxicity. NeuroMol Med 15(1):192–208
64. Cho N, Choi JH, Yang H, Jeong EJ, Lee KY, Kim YC, Sung SH (2012) Neuroprotective and
anti-inflammatory effects of flavonoids isolated from Rhus verniciflua in neuronal HT22 and
microglial BV2 cell lines. Food Chem Toxicol 50(6):1940–1945
65. Cho N, Lee KY, Huh J, Choi JH, Yang H, Jeong EJ, Kim HP, Sung SH (2013)
Cognitive-enhancing effects of Rhus verniciflua bark extract and its active flavonoids with
neuroprotective and anti-inflammatory activities. Food Chem Toxicol 58:355–361
66. Cho Y, Chung JH, Do HJ, Jeon HJ, Jin T, Shin MJ (2013) Effects of fisetin supplementation
on hepatic lipogenesis and glucose metabolism in Sprague–Dawley rats fed on a high fat diet.
Food Chem 139(1–4):720–727
67. Chen CM, Hsieh YH, Hwang JM, Jan HJ, Hsieh SC, Lin SH, Lai CY (2015) Fisetin
suppresses ADAM9 expression and inhibits invasion of glioma cancer cells through
increased phosphorylation of ERK1/2. Tumour Biol 36(5):3407–3415
68. Chen PY, Ho YR, Wu MJ, Huang SP, Chen PK, Tai MH, Ho CT, Yen JH (2015)
Cytoprotective effects of fisetin against hypoxia-induced cell death in PC12 cells. Food
Funct. 6(1):287–296
69. Prasath GS, Subramanian SP (2011) Modulatory effects of fisetin, a bioflavonoid, on
hyperglycemia by attenuating the key enzymes of carbohydrate metabolism in hepatic and
renal tissues in streptozotocin-induced diabetic rats. Eur J Pharmacol 668(3):492–496
70. Prasath GS, Subramanian SP (2014) Antihyperlipidemic effect of fisetin, a bioflavonoid of
strawberries, studied in streptozotocin-induced diabetic rats. J Biochem Mol Toxicol 28
(10):442–449
242 H.C. Pal et al.
71. Kim HJ, Kim SH, Yun JM (2012) Fisetin inhibits hyperglycemia-induced proinflammatory
cytokine production by epigenetic mechanisms. Evid Based Complement Alternat Med
2012:639469
72. Kan E, Kiliçkan E, Ayar A, Colak R (2014) Effects of two antioxidants; a-lipoic acid and
fisetin against diabetic cataract in mice. Int Ophthalmol [Epub ahead of print] PubMed
PMID: 25488016
73. Zhao X, Li XL, Liu X, Wang C, Zhou DS, Ma Q, Zhou WH, Hu ZY (2015) Antinociceptive
effects of fisetin against diabetic neuropathic pain in mice: engagement of antioxidant
mechanisms and spinal GABA(A) receptors. Pharmacol Res 102:286–297
74. Zhao X, Wang C, Cui WG, Ma Q, Zhou WH (2015) Fisetin exerts antihyperalgesic effect in a
mouse model of neuropathic pain: engagement of spinal serotonergic system. Sci Rep 5:9043
75. Jung CH, Kim H, Ahn J, Jeon TI, Lee DH, Ha TY (2013) Fisetin regulates obesity by
targeting mTORC1 signaling. J Nutr Biochem 24(8):1547–1554
76. Lee Y, Bae EJ (2013) Inhibition of mitotic clonal expansion mediates fisetin-exerted
prevention of adipocyte differentiation in 3T3-L1 cells. Arch Pharm Res 36(11):1377–1384
77. Jin T, Kim OY, Shin MJ, Choi EY, Lee SS, Han YS, Chung JH (2014) Fisetin up-regulates
the expression of adiponectin in 3T3-L1 adipocytes via the activation of silent mating type
information regulation 2 homologue 1 (SIRT1)-deacetylase and peroxisome
proliferator-activated receptors (PPARs). J Agric Food Chem 62(43):10468–10474
78. Kwon O, Eck P, Chen S, Corpe CP, Lee JH, Kruhlak M, Levine M (2007) Inhibition of the
intestinal glucose transporter GLUT2 by flavonoids. FASEB J 21(2):366–377
79. Jeon TI, Park JW, Ahn J, Jung CH, Ha TY (2013) Fisetin protects against hepatosteatosis in
mice by inhibiting miR-378. Mol Nutr Food Res 57(11):1931–1937
80. Lima LCF, Braga VA, do Socorro de França Silva M, Cruz JC, Sousa Santos SH, de Oliveira
Monteiro MM, Balarini CM (2015) Adipokines, diabetes and atherosclerosis: an
inflammatory association. Front Physiol 6:304
81. Viola J, Soehnlein O (2015) Atherosclerosis—a matter of unresolved inflammation. Semin
Immunol 27(3):184–193
82. Wong BW, Meredith A, Lin D, McManus BM (2012) The biological role of inflammation in
atherosclerosis. Can J Cardiol 28(6):631–641
83. Chistiakov DA, Bobryshev YV, Orekhov AN (2015) Neutrophil’s weapons in
atherosclerosis. Exp Mol Pathol 99(3):663–671
84. Pende A, Artom N, Bertolotto M, Montecucco F, Dallegri F (2015) Role of Neutrophils in
atherogenesis: an update. Eur J Clin Invest [Epub ahead of print]. doi:10.1111/eci.12566
85. Back M, Hansson GK (2015) Anti-inflammatory therapies for atherosclerosis. Nat Rev
Cardiol 12(4):199–211
86. Khan R, Spagnoli V, Tardif JC, L’Allier PL (2015) Novel anti-inflammatory therapies for the
treatment of atherosclerosis. Atherosclerosis. 240(2):497–509
87. Yamashita T, Sasaki N, Kasahara K, Hirata K (2015) Anti-inflammatory and
immune-modulatory therapies for preventing atherosclerotic cardiovascular disease.
J Cardiol 66(1):1–8
88. de Whalley CV, Rankin SM, Hoult JR, Jessup W, Leake DS (1990) Flavonoids inhibit the
oxidative modification of low density lipoproteins by macrophages. Biochem Pharmacol 39
(11):1743–1750
89. Podrez EA (2010) Anti-oxidant properties of high-density lipoprotein and atherosclerosis.
Clin Exp Pharmacol Physiol 37(7):719–725
90. Lian TW, Wang L, Lo YH, Huang IJ, Wu MJ (2008) Fisetin, morin and myricetin attenuate
CD36 expression and oxLDL uptake in U937-derived macrophages. Biochim Biophys Acta
1781(10):601–609
91. Podrez EA, Abu-Soud HM, Hazen SL (2000) Myeloperoxidase-generated oxidants and
atherosclerosis. Free Radic Biol Med 28(12):1717–1725
92. Chiang HM, Chan SY, Chu Y, Wen KC (2015) Fisetin ameliorated photodamage by
suppressing the mitogen-activated protein kinase/matrix metalloproteinase pathway and
nuclear factor-jB pathways. J Agric Food Chem 63(18):4551–4560
10 Fisetin and Its Role in Chronic Diseases 243
93. Seo SH, Jeong GS (2015) Fisetin inhibits TNF-a-induced inflammatory action and hydrogen
peroxide-induced oxidative damage in human keratinocyte HaCaT cells through
PI3 K/AKT/Nrf-2-mediated heme oxygenase-1 expression. Int Immunopharmacol 29
(2):246–253
94. Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M,
Marchetti MA, McArthur G, Ribas A, Roesch A, Hauschild A (2015) Melanoma. Nature
Reviews Disease Primers. Article number: 15003, Published online: 23 April 2015
95. Siegel RL, Miller KD, Jemal A (2015) Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29
96. Melnikova V, Bar-Eli M (2007) Inflammation and melanoma growth and metastasis: the role
of platelet-activating factor (PAF) and its receptor. Cancer Metastasis Rev 26(3–4):359–371
97. Melnikova VO, Bar-Eli M (2009) Inflammation and melanoma metastasis. Pigment Cell
Melanoma Res. 22(3):257–267
98. Richmond A, Yang J, Su Y (2009) The good and the bad of chemokines/chemokine
receptors in melanoma. Pigment Cell Melanoma Res 22(2):175–186
99. Dunn JH, Ellis LZ, Fujita M (2012) Inflammasomes as molecular mediators of inflammation
and cancer: potential role in melanoma. Cancer Lett 314(1):24–33
100. Syed DN, Lall RK, Chamcheu JC, Haidar O, Mukhtar H (2014) Involvement of ER stress
and activation of apoptotic pathways in fisetin induced cytotoxicity in human melanoma.
Arch Biochem Biophys 563:108–117
101. Syed DN, Chamcheu JC, Khan MI, Sechi M, Lall RK, Adhami VM, Mukhtar H (2014)
Fisetin inhibits human melanoma cell growth through direct binding to p70S6 K and mTOR:
findings from 3-D melanoma skin equivalents and computational modeling. Biochem
Pharmacol 89(3):349–360
102. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A (2015) Global cancer
statistics, 2012. CA Cancer J Clin 65(2):87–108
103. Haddad AQ, Venkateswaran V, Viswanathan L, Teahan SJ, Fleshner NE, Klotz LH (2006)
Novel antiproliferative flavonoids induce cell cycle arrest in human prostate cancer cell lines.
Prostate Cancer Prostatic Dis 9(1):68–76
104. Khan N, Afaq F, Syed DN, Mukhtar H (2008) Fisetin, a novel dietary flavonoid, causes
apoptosis and cell cycle arrest in human prostate cancer LNCaP cells. Carcinogenesis 29
(5):1049–1056
105. Mukhtar E, Adhami VM, Sechi M, Mukhtar H (2015) Dietary flavonoid fisetin binds to
b-tubulin and disrupts microtubule dynamics in prostate cancer cells. Cancer Lett 367
(2):173–183
106. Khan MI, Adhami VM, Lall RK, Sechi M, Joshi DC, Haidar OM, Syed DN, Siddiqui IA,
Chiu SY, Mukhtar H (2014) YB-1 expression promotes epithelial-to-mesenchymal transition
in prostate cancer that is inhibited by a small molecule fisetin. Oncotarget. 5(9):2462–2474
107. Lu X, Ji Jung, Cho HJ, Lim DY, Lee HS, Chun HS, Kwon DY, Park JH (2005) Fisetin
inhibits the activities of cyclin-dependent kinases leading to cell cycle arrest in HT-29 human
colon cancer cells. J Nutr 135(12):2884–2890
108. do Lim Y, Park JH (2009) Induction of p53 contributes to apoptosis of HCT-116 human
colon cancer cells induced by the dietary compound fisetin. Am J Physiol Gastrointest Liver
Physiol 296(5):G1060–G1068
109. Suh Y, Afaq F, Johnson JJ, Mukhtar H (2009) A plant flavonoid fisetin induces apoptosis in
colon cancer cells by inhibition of COX2 and Wnt/EGFR/NF-kappaB-signaling pathways.
Carcinogenesis 30(2):300–307
110. Yu SH, Yang PM, Peng CW, Yu YC, Chiu SJ (2011) Securin depletion sensitizes human
colon cancer cells to fisetin-induced apoptosis. Cancer Lett 300(1):96–104
111. Wu MS, Lien GS, Shen SC, Yang LY, Chen YC (2013) HSP90 inhibitors, geldanamycin and
radicicol, enhance fisetin-induced cytotoxicity via induction of apoptosis in human colonic
cancer cells. Evid Based Complement Alternat Med 2013:987612
112. Wu MS, Lien GS, Shen SC, Yang LY, Chen YC (2014) N-Acetyl-L-cysteine enhances
fisetin-induced cytotoxicity via induction of ROS-independent apoptosis in human colonic
cancer cells. Mol Carcinog 53(Suppl 1):E119–E129
244 H.C. Pal et al.
113. Cho WC, Kwan CK, Yau S, So PP, Poon PC, Au JS (2011) The role of inflammation in the
pathogenesis of lung cancer. Expert Opin Ther Targets 15(9):1127–1137
114. O’Callaghan DS, O’Donnell D, O’Connell F, O’Byrne KJ (2010) The role of inflammation in
the pathogenesis of non-small cell lung cancer. J Thorac Oncol 5(12):2024–2036
115. Bremnes RM, Al-Shibli K, Donnem T, Sirera R, Al-Saad S, Andersen S, Stenvold H,
Camps C, Busund LT (2011) The role of tumor-infiltrating immune cells and chronic
inflammation at the tumor site on cancer development, progression, and prognosis: emphasis
on non-small cell lung cancer. J Thorac Oncol 6(4):824–833
116. Gomes M, Teixeira AL, Coelho A, Araújo A, Medeiros R (2014) The role of inflammation in
lung cancer. Adv Exp Med Biol 816:1–23
117. Liao YC, Shih YW, Chao CH, Lee XY, Chiang TA (2009) Involvement of the ERK
signaling pathway in fisetin reduces invasion and migration in the human lung cancer cell
line A549. J Agric Food Chem 57(19):8933–8941
118. Kang KA, Piao MJ, Hyun JW (2015) Fisetin induces apoptosis in human nonsmall lung
cancer cells via a mitochondria-mediated pathway. Vitro Cell Dev Biol Anim 51(3):300–309
119. Ravichandran N, Suresh G, Ramesh B, Siva GV (2011) Fisetin, a novel flavonol attenuates
benzo(a)pyrene-induced lung carcinogenesis in Swiss albino mice. Food Chem Toxicol 49
(5):1141–1147
120. Ravichandran N, Suresh G, Ramesh B, Manikandan R, Choi YW, Vijaiyan Siva G (2014)
Fisetin modulates mitochondrial enzymes and apoptotic signals in benzo(a)pyrene-induced
lung cancer. Mol Cell Biochem 390(1–2):225–234
121. Touil YS, Seguin J, Scherman D, Chabot GG (2011) Improved antiangiogenic and
antitumour activity of the combination of the natural flavonoid fisetin and cyclophosphamide
in Lewis lung carcinoma-bearing mice. Cancer Chemother Pharmacol 68(2):445–455
122. Goh FY, Upton N, Guan S, Cheng C, Shanmugam MK, Sethi G, Leung BP, Wong WS
(2012) Fisetin, a bioactive flavonol, attenuates allergic airway inflammation through negative
regulation of NF-jB. Eur J Pharmacol 679(1–3):109–116
123. Wu MY, Hung SK, Fu SL (2011) Immunosuppressive effects of fisetin in ovalbumin-induced
asthma through inhibition of NF-jB activity. J Agric Food Chem 59(19):10496–10504
124. Feng G, Jiang ZY, Sun B, Fu J, Li TZ (2015) Fisetin alleviates lipopolysaccharide-induced
acute lung injury via TLR4-Mediated NF-jB signaling pathway in rats. Inflammation [Epub
ahead of print] PubMed PMID: 26272311
125. Higa S, Hirano T, Kotani M, Matsumoto M, Fujita A, Suemura M, Kawase I, Tanaka T
(2003) Fisetin, a flavonol, inhibits TH2-type cytokine production by activated human
basophils. J Allergy Clin Immunol 111(6):1299–1306
126. Hirano T, Higa S, Arimitsu J, Naka T, Shima Y, Ohshima S, Fujimoto M, Yamadori T,
Kawase I, Tanaka T (2004) Flavonoids such as luteolin, fisetin and apigenin areinhibitors of
interleukin-4 and interleukin-13 production by activated human basophils. Int Arch Allergy
Immunol 134(2):135–140
127. Morimoto Y, Yasuhara T, Sugimoto A, Inoue A, Hide I, Akiyama M, Nakata Y (2003)
Anti-allergic substances contained in the pollen of Cryptomeria japonica possess diverse
effects on the degranulation of RBL-2H3 cells. J Pharmacol Sci 92(3):291–295
128. Park HH, Lee S, Son HY, Park SB, Kim MS, Choi EJ, Singh TS, Ha JH, Lee MG, Kim JE,
Hyun MC, Kwon TK, Kim YH, Kim SH (2008) Flavonoids inhibit histamine release and
expression of proinflammatory cytokines in mast cells. Arch Pharm Res. 31(10):1303–1311
129. Nagai K, Takahashi Y, Mikami I, Fukusima T, Oike H, Kobori M (2009) The
hydroxyflavone, fisetin, suppresses mast cell activation induced by interaction with
activated T cell membranes. Br J Pharmacol 158(3):907–919
130. Kim JH, Kim MY, Kim JH, Cho JY (2015) Fisetin suppresses macrophage-mediated
inflammatory responses by blockade of Src and Syk. Biomol Ther (Seoul) 23(5):414–420
Chapter 11
Honokiol, an Active Compound
of Magnolia Plant, Inhibits Growth,
and Progression of Cancers of Different
Organs
Keywords Honokiol Ultraviolet radiation Cancer of different organs Cell
cycle regulation Inflammatory mediators Tumor cell migration
Abbreviations
BCC Basal cell carcinomas
CDK Cyclin dependent kinases
CHS Contact hypersensitivity
COX-2 Cyclooxygenase-2
EGFR Epidermal growth factor receptor
EMT Epithelial-mesenchymal transition
HNSCC Head and neck squamous cell carcinoma
IL Interleukin
iNOS Inducible nitric oxide synthase
MMP Matrix metalloproteinase
NF-jB Nuclear factor-kappa B
NSCLC Non-small cell lung cancer
PCNA Proliferating cell nuclear antigen
PG Prostaglandin
PGE2 Prostaglandin E2
SCC Squamous cell carcinomas
TNF-a Tumor necrosis factor-alpha
UVR Ultraviolet radiation
11.1 Introduction
therefore, in the recent past, honokiol achieved a great deal of research interest due
to its diverse biologic and pharmacologic activities that include antibacterial,
anti-inflammatory, anti-fungal, anti-oxidative, and anti-carcinogenic effects [1–8].
Chemically, honokiol is hydrophobic in nature but soluble in organic solvents, such
as acetone. Therefore, to avoid the use of organic solvents in some topical appli-
cations and formulation, which may cause deleterious effects, the research labo-
ratory of Dr. Katiyar has developed a topical formulation by mixing it in a
hydrophilic cream, and this cream-based topical formulation is ready and
easy-to-use for experimental purposes [8].
The protective and therapeutic effect of phytochemicals such as honokiol may be
associated with their antioxidant activity, as overproduction of reactive oxygen and
nitrogen species in the human body is involved in the pathogenesis of many chronic
diseases including cancer. To provide better understanding of the use of honokiol in
prevention and treatment of chronic diseases such lung cancer, prostate cancer, head
and neck cancer, gastric cancer, prostate cancer, urinary bladder cancer and neurob-
lastoma, etc., we are summarizing and explaining the investigations conducted in vitro
and in vivo animal models. It is well documented that many diseases occur or initiated
due to chronic and sustained inflammation in animals as well as in humans, which
includes cancer of several organs and aging processes, etc. Inflammation is a localized
reaction of tissue to infection, irritation, or other injury, e.g., exposure of the skin to
solar ultraviolet (UV) radiation. The key features of inflammation are redness or ery-
thema, warmth, swelling, pain, etc. Inflammation, considered as a necessary response to
clear viral infection, repair tissue insults and suppress tumor initiation and progression.
However, when inflammation is chronic and persists, diseases may develop, including
cancer. Importantly, inflammation involves in all the three stages of tumor develop-
ment: initiation, promotion/progression, and metastasis. During the initiation phase,
inflammation induces the release of a variety of cytokines and chemokines (mostly
pro-inflammatory) that promote the activation of inflammatory cells. These conditions
change the tissue microenvironment, which resulted in the favor of increased cell
survival and proliferation. Clinical and epidemiological evidences suggest a connection
between inflammation and a predisposition for the development of cancer. Here, we
summarize and discuss the therapeutic effects and mechanisms of action of honokiol
against cancers of some specific organs with particular emphasis on ultraviolet
(UV) radiation-induced skin cancer development.
Fig. 11.2 This generalized schematic diagram depicts the mechanism of UVR-induced inflam-
mation in the skin. UVR exposure induces inflammatory responses, including overexpression of
COX-2 and PGs production and reactive oxygen species (ROS) generation at early time points
(within few hours) of irradiation. Inflammatory mediators and ROS act as chemotactic factors and
stimulate the infiltration of leukocytes, particularly activated macrophages and neutrophils, at UV
irradiated skin site. Peak time of infiltration is in between 48 and 72 h after UV irradiation of the
skin. Activated infiltrating leukocytes are the major source of inflammatory mediators as well as
ROS. Topical treatment of the skin with honokiol inhibits UVR-induced effects both at 1st stage
(early stage), and 2nd stage (48–72 h after UV) through inhibition of leukocyte infiltration.
Inhibition of UVR-induced inflammatory mediators as well as ROS by honokiol treatment
contributes to the prevention of UVR-induced skin tumor development
PGE2 were significantly lower in the UVB-irradiated mouse skin, which was treated
with honokiol. Skin exposure to UV radiation induces infiltration of inflammatory
leukocytes in the skin, and most prominently are the activated macrophages and
neutrophils. These infiltrating leukocytes (majority of them are CD11b+ cell subset)
have a role in UV-induced immune suppression, and are the major source of inflam-
matory mediators, such as prostaglandins and pro-inflammatory cytokines at the
UV-exposed site, as demonstrated in Fig. 11.2. In addition, these infiltrating CD11b+
leukocytes have been shown to have suppressive effects on immune system. UVB
irradiation also induces production of pro-inflammatory cytokines, such as tumor
necrosis factor-alpha (TNF-a), interleukin (IL)-1b, IL-6, etc., in the skin. Topical
treatment of mouse skin with honokiol resulted in a significant reduction in
UVB-induced production of TNF-a, IL-1b, IL-6 as compared to non-honokiol-treated
UVB-exposed mouse skin.
250 R. Prasad and S.K. Katiyar
Non-melanoma skin cancer (NMSC) is the most common cancer in the United States.
The majority of NMSCs is environmentally induced and caused by excessive exposure
to solar UVB radiation which induces inflammation, oxidative stress, suppression of
immune system, and DNA damage. NMSC is composed of two types of skin cancer;
basal cell carcinomas (BCCs), and squamous cell carcinomas (SCCs). The incidence of
SCC has increased approximately 200 % over the past three decades, and represents
about 20 % of NMSC [19, 20]. As topical treatment of honokiol prevents
UVB-induced inflammation and their mediators in the mouse skin, the effect of hon-
okiol was assessed against UVB-induced skin tumor development in SKH-1 hairless
mouse model [8]. To induce tumors, mice were exposed to UVB radiation
(180 mJ/cm2) 3 times a week for 24 weeks. It was observed that topical treatment of
honokiol significantly inhibits UVB-induced initiation and progression of skin tumors
[8]. Honokiol treatment also increased the latency period of skin tumor development.
The tumor multiplicity and tumor size were significantly reduced in the group of
UVB-irradiated mice that were treated with honokiol than in the control group of mice
that were UVB-irradiated but not treated with honokiol. When data were compared in
terms of average tumor volume/tumor bearing mouse between honokiol-treated and
non-honokiol-treated groups, a significant reduction was observed after the treatment of
honokiol. Similar observations were also noted by Chilampalli et al. [21] wherein
chemopreventive effect of honokiol was determined on UVB-induced skin carcino-
genesis. However, these investigators have determined the chemopreventive effect of
topical treatment of honokiol at lower dose (30 lg/200 ml acetone/mouse) as well as
lower dose of UVB exposure (30 mJ/cm2). This study also concluded that honokiol
treatment affords photoprotection in terms of tumor multiplicity. In addition to the
inhibition of skin tumor development in UVB-exposed mouse skin, the malignant
progression of papillomas to carcinomas were also significantly prevented by the
11 Honokiol, an Active Compound of Magnolia Plant … 251
topical treatment of honokiol in mice. Histochemical analysis revealed that most of the
carcinomas were identified as kerato-acanthomas and squamous cell carcinomas [8].
such as PI3K/Akt [27, 28]. Studies revealed that the levels of both the catalytic
(p110) and regulatory (p85) subunits of PI3K were enhanced in the UVB-induced
skin tumors as compared with the non-UVB-exposed normal mouse skin; however,
the levels of the p85 and p110 subunits were greatly reduced in the UV-induced
skin tumors from honokiol-treated mice compared to the skin tumors of
non-honokiol-treated mice. Most of the biological effects of PI3K are mediated
through the activation of the downstream target Akt. Akt is a serine/threonine
kinase, which has been identified as an important component of pro-survival sig-
naling pathways [29]. It was observed that the treatment of honokiol resulted in
reduction of UVB-induced phosphorylation of Akt (Ser473) as compared to the skin
tumors of non-honokiol-treated group. Further, cell survival signals have been
associated with cellular proliferation and carcinogenesis [30–32]. The skin tumors
augment UVB radiation-induced activation of PI3K and phospho-Akt as compared
to the non-UVB-exposed mouse skin. The PI3K/Akt signaling pathway regulates
the activity of the transcriptional factor, NF-jB, which in turn known to regulate
several well-known markers of tumor promotion and tumor cell proliferation, e.g.,
COX-2, inducible nitric oxide synthase (iNOS) and PCNA [33, 34]. Thus, the
inhibition of PI3K/p-Akt pathway in skin tumors by honokiol may have a role in
inhibition of UVB-induced skin tumor growth in mouse model.
Urinary bladder cancer is one of the most common urogenital malignant cancer
with an estimated 74,690 new cases and 15,580 deaths occurring in USA during
2014 [70]. Recent studies reported that cancer stem cells might account for
256 R. Prasad and S.K. Katiyar
chemotherapy failure, which are enriched after therapy and have the ability to
generate all types of differentiated cells to repopulate tumors and eventually lead to
metastasis [71–73]. Histone modifications through polycomb repressive complexes
also play an essential role for normal and malignant cell stemness maintenance.
Deregulation of Enhancer of Zeste Homologue 2 (EZH2), an important component
of polycomb repressive complex 2, is frequently detected in a variety of cancer
along with urinary bladder cancer [74–76]. Activation EZH2 specifically represses
the transcription of differentiation-related genes throughout the cell cycle to
maintain the stemness of cells, and this make it a promising therapeutic target for
cancer. Zhang et al. [77] have found that depletion of EZH2 by honokiol treatment
inhibited cell proliferation and clonogenicity of urinary bladder cancer cells.
Prostate cancer is the most frequently diagnosed cancer and the second leading
cause of cancer-related death in males after skin cancer in economically developed
countries [70]. The major cause of mortality in prostate cancer is associated with
metastasis. Approximately, 90 % of deaths from solid tumors are caused by
metastasis [78]. Studies have shown that honokiol induces autophagy in cancer
cells [79, 80]. An induction of autophagy with different functional consequences
has been described for a number of structurally divergent naturally occurring
anticancer agents. Studies have implicated that mTOR is a negative regulator of
autophagy. A study published by Hahm et al. [48] reported the suppression of
mTOR and Akt phosphorylation by honokiol treatment in prostate cancer cells
(PC-3). An induction of apoptosis by natural agents is significantly attenuated by
antioxidants because of reduced activation of multi domain Bcl-2 family member
Bax. Hahm et al. [48] also found that treatment of honokiol induced apoptosis in
PC-3 and LNCaP cells, which was associated with induction of Bax and Bak, and
claimed that honokiol may be effective in prevention and therapy of prostate cancer.
Gastric cancer is also a leading cause of cancer-related death worldwide, and the
majority of patients exhibit a high incidence of lymph node metastases. Emerging
evidence suggests that EMT leads to increased tumor formation, tissue invasiveness,
11 Honokiol, an Active Compound of Magnolia Plant … 257
and tumor dissemination [81]. Recent studies have shown a close relation between
EMT and gastric cancer progression. A number of signaling pathways, including
developmental transcriptional factors, are involved in regulating the motile-invasion
phenotype of tumor cells [82]. The decreased expression of epithelial marker
E-cadherin in gastric cancer has established the potential role of EMT in gastric
cancer metastasis [83, 84]. Tumor progression locus 2 (Tpl2) is a serine-threonine
kinase, regulates the activation of the mitogen-activated protein kinase, and critically
involved in inflammation, oncogenic events, and tumor progression [85, 86]. Pan
et al. [87] have shown that honokiol treatment regulates Tpl2 mediated mesenchy-
mal markers and significantly down regulates Snail, vimentin, N-cadherin expres-
sion, and upregulates cytokeratin-18 and E-cadherin expression. Tumor growth in
gastric cancer has been associated with Tpl2 [88, 89]. Pan et al. [87] also reported
that honokiol treatment reduces growth of gastric tumor through an inhibition of
Tpl2 expression. Honokiol significantly inhibited tumor angiogenesis as indicated
by reduced microvessel density in tumor mass [87]. Cell cycle regulation is an
important regulatory mechanism to control cell growth, and activation of p53, a
tumor suppressor protein, is involved in the regulation of cell cycle arrest and
apoptosis. The phosphorylation of cell cycle regulatory proteins is involved in
arresting effect of gastric carcinoma cells on the cell cycle at G2/M phase [90, 91].
Yen et al. [92] investigated the anticancer mechanism of honokiol in human gastric
carcinoma using MGC-803 cells and investigated that honokiol induces apoptosis in
gastric carcinoma cells, and the underlying mechanism was mediated through
downregulation of CDC2/cdc25C and upregulation of p53. These studies provide
evidence of anticancer activity of honokiol against human gastric carcinoma.
Lung cancer is a major cause of cancer-related deaths in the United States as well as
worldwide each year and thus have a tremendous impact on human health and
health care expenditures. Non-small-cell lung cancer (NSCLC) accounts for
approximately 80 % of all types of lung cancer. COX-2 is frequently over express
in lung cancer and associated with excessive production of PGE2, which promotes
tumor cell survival, invasion and metastasis [93–96]. Therefore, COX-2 inhibitors
have shown potential in treatment of lung cancer. Singh and Katiyar [97] reported
the use of honokiol as an inhibitor of COX-2 expression to inhibit migratory
potential of lung cancer cells and supported their findings by the evidence that
treatment of the NSCLC cells with celecoxib, a potent COX-2 inhibitor, resulted in
a reduction in cell migration. The b-catenin signaling is downstream target of
258 R. Prasad and S.K. Katiyar
COX-2 and played important roles in tumor progression as well as cell migration.
b-catenin forms a dynamic link between E-cadherin and cytoskeleton [98, 99], and
this cell-to-cell adhesion may prevent the migration of tumor cells. In contrast, the
breaking of cell-to-cell adhesion due to activation of b-catenin and its nuclear
accumulation may increase the migration potential of tumor cells. Singh and
Katiyar [97] also reported that honokiol induced degradation of b-catenin or
reduces nuclear accumulation, which leads to inhibition of lung cancer cell
migration.
Pancreatic cancer is one of the most lethal malignancies with increasing incidence in
the United States [33]. Due to its asymptomatic progression, pancreatic cancer is
diagnosed at later stage, when it has already metastasized or locally advanced. The
NF-jB is constitutively activated in a variety of hematologic and solid malignancies,
including pancreatic cancer and controls the expression of an array of genes involved in
cell proliferation and survival through direct and indirect mechanisms [100]. Arora
et al. [101, 102] examined the effect of honokiol against pancreatic cancer and reported
that honokiol showed growth inhibitory potential for pancreatic cancer lines (such as,
Miapaca and PANC-1), which may result of cell cycle arrest and induction of apop-
tosis. Furthermore, to explore the underlying mechanism, these authors have tested the
effect of honokiol on NF-jB signaling in this system. Treatment of honokiol to pan-
creatic cancer cells inhibited transcriptional activity of NF-jB, and decrease protein
expression in the nuclear fraction and suppresses constitutive activation of NF-jB in
pancreatic cancer cells and thus induce cell death.
Fig. 11.3 The schematic diagram reflects the preventative and therapeutic effect of honokiol on
cancers of different organs. Various molecular and cellular targets in cancers of different organs are
affected by the treatment of honokiol in vitro and in vivo models. Inflammation and inflammatory
mediators are the main targets of honokiol in prevention or treatment of cancers. Underline words
indicate the name of organ-specific cancer
Acknowledgments The work reported from Dr. Katiyar’s research laboratory was financially
supported from the funds from National Cancer Institute/NIH (CA183869) and Veterans
Administration Merit Review Award (1I01BX001410). The content of this communication does
not necessarily reflect the views or policies of the funding agencies.
References
1. Li TSC (2002) Chinese and related North American herbs: Phytopharmacology and
therapeutic values. CRC Press, Boca Raton, FL
2. Hahm ER, Arlotti JA, Marynowski SW, Singh SV (2008) Honokiol, a constituent of oriental
medicinal herb magnolia officinalis, inhibits growth of PC-3 xenografts in vivo in association
with apoptosis induction. Clin Cancer Res 14:1248–1257
260 R. Prasad and S.K. Katiyar
23. Graña X, Reddy EP (1995) Cell cycle control in mammalian cells: role of cyclins, cyclin
dependent kinases (CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors
(CKIs). Oncogene 11:211–219
24. Sherr CJ, Roberts JM (1999) CDK inhibitors: positive and negative regulators of G1-phase
progression. Genes Dev 13:1501–1512
25. Morgan DO (1995) Principles of CDK regulation. Nature 374:131–134
26. Fotedar R, Bendjennat M, Fotedar A (2004) Role of p21WAF1 in the cellular response to
UV. Cell Cycle 3:134–137
27. Luo J, Manning BD, Cantley LC (2003) Targeting the PI3K-Akt pathway in human cancer:
rationale and promise. Cancer Cell 4:257–262
28. Nomura M, Kaji A, Ma WY, Zhong S, Liu G, Bowden GT, Miyamoto KI, Dong Z (2001)
Mitogen- and stress-activated protein kinase 1 mediates activation of Akt by ultraviolet B
irradiation. J Biol Chem 276:25558–25567
29. Downward J (1998) Mechanisms and consequences of activation of protein kinase B/Akt.
Curr Opin Cell Biol 10:262–267
30. Tyrrell RM (1996) Activation of mammalian gene expression by the UV component of
sunlight-from models to reality. BioEssays 18:139–148
31. Osaki M, Kase S, Adachi K, Takeda A, Hashimoto K, Ito H (2004) Inhibition of the
PI3K-Akt signaling pathway enhances the sensitivity of Fas-mediated apoptosis in human
gastric carcinoma cell line, MKN-45. J Cancer Res Clin Oncol 130:8–14
32. Saleem M, Afaq F, Adhami VM, Mukhtar H (2004) Lupeol modulates NF-kappaB and
PI3K/Akt pathways and inhibits skin cancer in CD-1 mice. Oncogene 23:5203–5214
33. Carpenter CL, Cantley LC (1996) Phosphoinositide kinases. Curr Opin Cell Biol 8:153–158
34. Callejas NA, Casado M, Bosca L, Martin-Sanz P (1999) Requirement of nuclear factor
kappaB for the constitutive expression of nitric oxide synthase-2 and cyclooxygenase-2 in rat
trophoblasts. J Cell Sci 18:3147–3155
35. American Cancer Society. Cancer facts and figures. http://www.cancer.org/research/
cancerfactsfigures/. Accessed July 2014
36. Lewis EM, Sergeant S, Ledford B, Stull N, Dinauer MC, McPhail LC (2010)
Phosphorylation of p22phox on threonine 147 enhances NADPH oxidase activity by
promoting p47phox binding. J Biol Chem 285:2959–2967
37. Sheppard FR, Kelher MR, Moore EE, McLaughlin NJ, Banerjee A, Silliman CC (2005)
Structural organization of the neutrophil NADPH oxidase: phosphorylation and translocation
during priming and activation. J Leukoc Biol 78:1025–1042
38. Prasad R, Kappes JC, Katiyar SK (2016) Inhibition of NADPH oxidase 1 activity and
blocking the binding of cytosolic and membrane-bound proteins by honokiol inhibit
migratory potential of melanoma cells. Oncotarget 7:7899–7912
39. Ambasta RK, Kumar P, Griendling KK, Schmidt HH, Busse R, Brandes RP (2004) Direct
interaction of the novel Nox proteins with p22phox is required for the formation of a
functionally active NADPH oxidase. J Biol Chem 279:45935–45941
40. Sumimoto H, Hata K, Mizuki K, Ito T, Kage Y, Sakaki Y, Fukumaki Y, Nakamura M,
Takeshige K (1996) Assembly and activation of the phagocyte NADPH oxidase. Specific
interaction of the N-terminal Src homology 3 domain of p47phox with p22phox is required for
activation of the NADPH oxidase. J Biol Chem 271:22152–22158
41. Hoehner JC, Gestblom C, Hedborg F, Sandstedt B, Olsen L, Pahlman S (1996) A
developmental model of neuroblastoma: differentiating stroma-poor tumors’ progress along
an extra-adrenal chromaffin lineage. Lab Invest 75:659–675
42. Park JR, Eggert A, Caron H (2008) Neuroblastoma: biology, prognosis, and treatment.
Pediatr Clin North Am 55:97–120
43. Zage PE, Kletzel M, Murray K, Marcus R, Castleberry R, Zhang Y, London WB,
Kretschmar C, Children’s Oncology Group (2008) Outcomes of the POG 9340/9341/9342
trials for children with high-risk neuroblastoma: a report from the Children’s Oncology
Group. Pediatr Blood Cancer 51:747–753
262 R. Prasad and S.K. Katiyar
44. Lin JW, Chen JT, Hong CY, Lin YL, Wang KT, Yao CJ, Lai GM, Chen RM (2012)
Honokiol traverses the blood-brain barrier and induces apoptosis of neuroblastoma cells via
an intrinsic Bax-mitochondrion-cytochrome c-caspase protease pathway. Neuro Oncol
14:302–314
45. Yeh PS, Wang W, Chang YA, Lin CJ, Wang JJ, Chen RM (2016) Honokiol induces
autophagy of neuroblastoma cells through activating the PI3K/Akt/mTOR and endoplasmic
reticular stress/ERK1/2 signaling pathways and suppressing cell migration. Cancer Lett
370:66–77
46. Lv X, Liu F, Shang Y, Chen SZ (2015) Honokiol exhibits enhanced antitumor effects with
chloroquine by inducing cell death and inhibiting autophagy in human non-small cell lung
cancer cells. Oncol Rep 34:1289–1300
47. Kaushik G, Venugopal A, Ramamoorthy P, Standing D, Subramaniam D, Umar S,
Jensen RA, Anant S, Mammen JM (2014) Honokiol inhibits melanoma stem cells by
targeting notch signaling. Mol Carcinog 54:1710–1721
48. Hahm ER, Sakao K, Singh SV (2014) Honokiol activates reactive oxygen species mediated
cytoprotective autophagy in human prostate cancer cells. Prostate 74:1209–1221
49. Guo YB, Bao XJ, Xu SB, Zhang XD, Liu HY (2015) Honokiol induces cell cycle arrest and
apoptosis via p53 activation in H4 human neuroglioma cells. Int J Clin Exp Med 15:7168–
7175
50. Kim KC, Guan KL (2015) mTOR: a pharmacologic target for autophagy regulation. J Clin
Invest 125:25–32
51. Hunter KD, Parkinson EK, Harrison PR (2005) Profiling early head and neck cancer. Nat
Rev Cancer 5:127–135
52. Casiglia J, Woo SB (2001) A comprehensive review of oral cancer. Gen Dent 49:72–82
53. Grandis JR, Melhem MF, Barnes EL, Tweardy DJ (1996) Quantitative immunohistochemical
analysis of transforming growth factor-a and epidermal growth factor receptor in patients
with squamous cell carcinoma of the head and neck. Cancer 78:1284–1292
54. He Y, Zeng Q, Drenning SD, Melhem MF, Tweardy DJ, Huang L, Grandis JR (1998)
Inhibition of human squamous cell carcinoma growth in vivo by epidermal growth factor
receptor antisense RNA transcribed from the U6 promoter. J Natl Cancer Inst 90:1080–1087
55. Grandis JR, Melhem MF, Gooding WE, Day R, Holst VA, Wagener MM, Drenning SD,
Tweardy DJ (1998) Levels of TGF-a and EGFR protein in head and neck squamous cell
carcinoma and patient survival. J Natl Cancer Inst 90:824–832
56. Singh T, Gupta NA, Xu S, Prasad R, Velu SE, Katiyar SK (2015) Honokiol inhibits the
growth of head and neck squamous cell carcinoma by targeting epidermal growth factor
receptor. Oncotarget 6:21268–21282
57. Crowell JA, Steele VE, Sigman CC, Fay JR (2003) Is inducible nitric oxide synthase a target
for chemoprevention? Mol Cancer Ther 2:815–823
58. Choi BD, Jeong SJ, Wang G, Park JJ, Lim DS, Kim BH, Cho YI, Kim CS, Jeong MJ (2011)
Secretory leukocyte protease inhibitor is associated with MMP-2 and MMP-9 to promote
migration and invasion in SNU638 gastric cancer cells. Int J Mol Med 28:527–534
59. Cho JH, Jeon YJ, Park SM, Shin JC, Lee TH, Jung S, Park H, Ryu J, Chen H, Dong Z,
Shim JH, Chae JI (2015) Multifunctional effects of honokiol as an anti-inflammatory and
anticancer drug in human oral squamous cancer cells and xenograft. Biomaterials 53:274–
284
60. Higo T, Hattori M, Nakamura T, Natsume T, Michikawa T, Mikoshiba K (2005) Subtype-specific
and ER lumenal environment-dependent regulation of inositol1,4,5-trisphosphate receptor type 1
by ERp44. Cell 120:85–98
61. Armstrong JS (2006) Mitochondrial membrane permeabilization: the sine qua non for cell
death. BioEssays 28:253–260
62. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F,
Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA (2008)
The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell
133:704–715
11 Honokiol, an Active Compound of Magnolia Plant … 263
63. Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S, Puisieux A (2008) Generation of
breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE 3:e2888
64. Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, Haluska P,
Ingle JN, Hartmann LC, Manjili MH, Radisky DC, Ferrone S, Knutson KL (2009)
Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem
cells. Cancer Res 69:2887–2895
65. Avtanski DB, Nagalingam A, Bonner MY, Arbiser JL, Saxena NK, Sharma D (2015)
Honokiol activates LKB1-miR-34a axis and antagonizes the oncogenic actions of leptin in
breast cancer. Oncotarget 6:29947–29962
66. Shen Z, Wen XF, Lan F, Shen ZZ, Shao ZM (2002) The tumor suppressor gene LKB1 is
associated with prognosis in human breast carcinoma. Clin Cancer Res 8:2085–2090
67. Li J, Liu J, Li P, Mao X, Li W, Yang J, Liu P (2014) Loss of LKB1 disrupts breast epithelial
cell polarity and promotes breast cancer metastasis and invasion. J Exp Clin Cancer Res
33:70
68. Roy BC, Kohno T, Iwakawa R, Moriguchi T, Kiyono T, Morishita K, Sanchez-Cespedes M,
Akiyama T, Yokota J (2010) Involvement of LKB1 in epithelial-mesenchymal transition
(EMT) of human lung cancer cells. Lung Cancer 70:136–145
69. Hermeking H (2010) The miR-34 family in cancer and apoptosis. Cell Death Differ 17:193–
199
70. Siegel R, Ma J, Zou Z, Jemal A (2014) Cancer statistics, 2014. CA Cancer J Clin 64:9–29
71. Galluzzi L, Vitale I, Michels J, Brenner C, Szabadkai G, Harel-Bellan A, Castedo M,
Kroemer G (2014) Systems biology of cisplatin resistance: past, present and future. Cell
Death Dis 5:e1257
72. Magee JA, Piskounova E, Morrison SJ (2012) Cancer stem cells: impact, heterogeneity, and
uncertainty. Cancer Cell 21:283–296
73. Valent P, Bonnet D, De Maria R, Lapidot T, Copland M, Melo JV, Chomienne C,
Ishikawa F, Schuringa JJ, Stassi G, Huntly B, Herrmann H, Soulier J, Roesch A,
Schuurhuis GJ, Wohrer S, Arock M, Zuber J, Cerny-Reiterer S, Johnsen HE, Andreeff M,
Eaves C (2012) Cancer stem cell definitions and terminology: the devil is in the details. Nat
Rev Cancer 12:767–775
74. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Sinha CK, Sanda MG, Ghosh D,
Pienta KJ, Sewalt RG, Otte AP, Rubin MA, Chinnaiyan AM (2002) The polycomb group
protein EZH2 is involved in progression of prostate cancer. Nature 419:624–629
75. Huqun Ishikawa R, Zhang J, Miyazawa H, Goto Y, Shimizu Y, Hagiwara K, Koyama N
(2012) Enhancer of zeste homolog 2 is a novel prognostic biomarker in nonsmall cell lung
cancer. Cancer 118:1599–1606
76. Raman JD, Mongan NP, Tickoo SK, Boorjian SA, Scherr DS, Gudas LJ (2005) Increased
expression of the polycomb group gene, EZH2, in transitional cell carcinoma of the bladder.
Clin Cancer Res 11:8570–8576
77. Zhang Q, Zhao W, Ye C, Zhuang J, Chang C, Li Y, Huang X, Shen L, Li Y, Cui Y, Song J,
Shen B, Eliaz I, Huang R, Ying H, Guo H, Yan J (2015) Honokiol inhibits bladder tumor
growth by suppressing EZH2/miR-143 axis. Oncotarget 6:37335–37348
78. Gupta GP, Massague J (2006) Cancer metastasis: building a framework. Cell 4:679–695
79. Chang KH, Yan MD, Yao CJ, Lin PC, Lai GM (2013) Honokiol-induced apoptosis and
autophagy in glioblastoma multiforme cells. Oncol Lett 6:1435–1438
80. Kaushik G, Ramalingam S, Subramaniam D, Rangarajan P, Protti P, Rammamoorthy P,
Anant S, Mammen JM (2012) Honokiol induces cytotoxic and cytostatic effects in malignant
melanoma cancer cells. Am J Surg 204:868–873
81. De Craene B, Berx G (2013) Regulatory networks defining EMT during cancer initiation and
progression. Nat Rev Cancer 13:97–110
264 R. Prasad and S.K. Katiyar
82. Rosivatz E, Becker I, Specht K, Fricke E, Luber B, Busch R, Höfler H, Becker KF (2002)
Differential expression of the epithelial-mesenchymal transition regulators snail, SIP1, and
twist in gastric cancer. Am J Pathol 161:1881–1891
83. Liu WF, Ji SR, Sun JJ, Zhang Y, Liu ZY, Liang AB, Zeng HZ (2012) CD146 expression
correlates with epithelial-mesenchymal transition markers and a poor prognosis in gastric
cancer. Int J Mol Sci 13:6399–6406
84. Zhong XY, Zhang LH, Jia SQ, Shi T, Niu ZJ, Du H, Zhang GG, Hu Y, Lu AP, Li JY, Ji JF
(2008) Positive association of up-regulated Cripto-1 and down-regulated E-cadherin with
tumour progression and poor prognosis in gastric cancer. Histopathology 52:560–568
85. Vougioukalaki M, Kanellis DC, Gkouskou K, Eliopoulos AG (2011) Tpl2 kinase signal
transduction in inflammation and cancer. Cancer Lett 304:80–89
86. Ohara R, Hirota S, Onoue H, Nomura S, Kitamura Y, Toyoshima K (1995) Identification of
the cells expressing cot proto-oncogene mRNA. J Cell Sci 108:97–103
87. Pan HC, Lai DW, Lan KH, Shen CC, Wu SM, Chiu CS, Wang KB, Sheu ML (2013)
Honokiol thwarts gastric tumor growth and peritoneal dissemination by inhibiting Tpl2 in an
orthotopic model. Carcinogenesis 34:2568–2579
88. Li YL, Vergne J, Torchet C, Maurel MC (2009) In vitro selection of adenine-dependent
ribozyme against Tpl2/Cot oncogene. FEBS J 276:303–314
89. Perfield JW 2nd, Lee Y, Shulman GI, Samuel VT, Jurczak MJ, Chang E, Xie C, Tsichlis PN,
Obin MS, Greenberg AS (2011) Tumor progression locus 2 (TPL2) regulates
obesity-associated inflammation and insulin resistance. Diabetes 60:1168–1176
90. Yunlan L, Juan Z (2014) Qingshan L (2014) Antitumor activity of di-n-butyl-
(2,6-difluorobenzohydrox-amato)tin (IV) against human gastric carcino-ma SGC-7901
cells via G2/M cell cycle arrest and cell apoptosis. PLoS ONE 9:e90793
91. Su CC (2014) Tanshinone IIA inhibits gastric carcino-ma AGS cells through increasing
p-p38, p-JNK and p53 but reducing p-ERK, CDC2 and cyclin B1 expression. Anticancer Res
34:7097–7110
92. Yan B, Peng ZY (2015) Honokiol induces cell cycle arrest and apoptosis in human gastric
carcinoma MGC-803 cell line. Int J Clin Exp Med 8:5454–5461
93. Huang M, Stolina M, Sharma S, Mao JT, Zhu L, Miller PW, Wollman J, Herschman H,
Dubinett SM (1998) Non-small cell lung cancer cyclooxygenase-2-dependent regulation of
cytokine balance in lymphocytes and macrophages: up-regulation of interleukin 10 and
down-regulation of interleukin 12 production. Cancer Res 58:1208–1216
94. Hida T, Yatabe Y, Achiwa H, Muramatsu H, Kozaki K, Nakamura S, Ogawa M,
Mitsudomi T, Sugiura T, Takahashi T (1998) Increased expression of cyclooxygenase 2
occurs frequently in human lung cancers, specifically in adenocarcinomas. Cancer Res
58:3761–3764
95. Wolff H, Saukkonen K, Anttila S, Karjalainen A, Vainio H, Ristimäki A (1998) Expression
of cyclooxygenase-2 in human lung carcinoma. Cancer Res 58:4997–5001
96. Hosomi Y, Yokose T, Hirose Y, Nakajima R, Nagai K, Nishiwaki Y, Ochiai A (2000)
Increased cyclooxygenase 2 (COX-2) expression occurs frequently in precursor lesions of
human adenocarcinoma of the lung. Lung Cancer 30:73–81
97. Singh T, Katiyar SK (2013) Honokiol inhibits non-small cell lung cancer cell migration by
targeting PGE2-mediated activation of b-catenin signaling. PLoS ONE 8:e60749
98. Tuynman JB, Vermeulen L, Boon EM, Kemper K, Zwinderman AH, Peppelenbosch MP,
Richel DJ (2008) Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt
activity in colon cancer. Cancer Res 68:1213–1220
99. Shlomo H, Simon JA (2008) A small-molecule inhibitor of Tcf/beta-catenin signaling
down-regulates PPARgamma and PPARdelta activities. Mol Cancer Ther 7:521–529
11 Honokiol, an Active Compound of Magnolia Plant … 265
100. Wharry CE, Haines KM, Carroll RG, May MJ (2009) Constitutive noncanonical NF-kappaB
signaling in pancreatic cancer cells. Cancer Biol Ther 8:1567–1576
101. Arora S, Bhardwaj A, Srivastava SK, Singh S, McClellan S, Wang B, Singh AP (2011)
Honokiol arrests cell cycle, induces apoptosis, and potentiates the cytotoxic effect of
gemcitabine in human pancreatic cancer cells. PLoS ONE 6:e21573
102. Arora S, Singh S, Piazza GA, Contreras CM, Panyam J, Singh AP (2012) Honokiol: a novel
natural agent for cancer prevention and therapy. Curr Mol Med 12:1244–1252
Chapter 12
Celastrol and Its Role
in Controlling Chronic Diseases
Keywords Celastrol Inflammation Autoimmune diseases Neurodegenerative
diseases Metabolic disorders
Immune modulation Natural products
Traditional Chinese medicine
12.1 Introduction
Fig. 12.1 Molecular structure of celastrol. Celastrol is a pentacyclic triterpenoid with a molecular
weight 450.6 and molecular formula C29H38O4. It belongs to a small class of organic compounds
known as quinone methides. Celastrol has an acidic group at one end and a phenolic quinone at the
other end
12 Celastrol and Its Role in Controlling Chronic Diseases 269
modifications within those proteins [6, 16–18]. Apparently, this is one of the
mechanisms by which celastrol can affect biological functions of proteins. Celastrol
is also known as tripterine/tripterin, but the name celastrol is commonly used.
For RA, using the rat adjuvant arthritis (AA) model, mouse collagen-induced
arthritis (CIA) model and fibroblast-like synoviocytes from RA patients (RA-FLS)
culture model, celastrol has been shown to reduce the severity of clinical and
histopathological features of arthritis, as well as to modulate the production of
pro-inflammatory cytokines and chemokines [8, 9, 19], to reset the T helper 17
(Th17)/T regulatory (Treg) cell balance to facilitate the suppression of arthritis [20],
and to afford protection against bone damage [8, 9, 19] (Table 12.1A). Celastrol
also inhibits RA-FLS invasion and protects against bone and cartilage damage [21].
For multiple sclerosis (MS), celastrol is shown to modulate Th17 responses, to shift
Th1 responses toward Th2, and to increase the production of anti-inflammatory
cytokines in the experimental autoimmune encephalomyelitis (EAE) model of MS
[10, 22]. For SLE (also known as lupus), celastrol treatment decreases transforming
growth factor (TGF)-b production, urine protein excretion, and serum autoantibody
levels in BW F1 and BALB/c mouse models of SLE [23–25]. For ulcerative colitis,
using the mouse dextran sulfate sodium (DSS)-induced colitis model, celastrol has
been shown to modulate oxidative stress, inflammatory cytokines and intestinal
homeostasis [11]. For asthma and other hypersensitivity reactions, celastrol inhibits
histamine and eotaxin production and other mediators involved in hypersensitivity
reactions [26–29]. The main mediators and pathways targeted by celastrol in
above-mentioned diseases are described in Table 12.1A.
270 S.H. Venkatesha and K.D. Moudgil
Celastrol can inhibit platelet activation [40], prevent atherosclerotic plaque size in
apo E-deficient mice [41], and decrease ratio of the plaque area and the arterial wall
cross-section area in a rabbit model of carotid atherosclerosis [41], thus revealing
the anti-atherosclerosis effect of this natural triterpene (Table 12.1C). Recently,
celastrol has been reported to be a leptin sensitizer, whose effects are manifest as
12 Celastrol and Its Role in Controlling Chronic Diseases 273
reduced intake of food, increased energy expenditure, and weight loss, and thereby
it may potentially serve as an anti-obesity agent [42]. Celastrol is also effective in
improving insulin resistance and limiting renal injury in a mouse model of type 2
diabetes (T2D) [43]. Furthermore, celastrol can modulate human immunodeficiency
virus (HIV) 1-transactivator of transcription (Tat)-induced inflammatory responses
in astrocytes in vitro by inhibiting the activation of various signaling pathways as
well as the expression of pro-inflammatory chemokines and adhesion molecules
such as intracellular adhesion molecule-1 (ICAM-1)/vascular cell adhesion
molecule-1 (VCAM-1) [44]. Celastrol has also been reported to target defined
functional components of pathogens such as the HIV-Tat [18] to inhibit the tran-
scription and replication of that virus, and the enzyme enoyl-acyl carrier protein
reductase of Plasmodium falciparum [45], which is a drug target for this malaria
parasite. However, because of the limited scope of this article, we have not elab-
orated further on direct effects of celastrol on various pathogens.
Fig. 12.2 Cell signaling pathways and other disease-related processes modulated by celastrol for
the control of chronic diseases. A schematic representation of the cell signaling pathways that are
regulated by celastrol is depicted here. These pathways are activated in response to diverse stimuli
induced by specific ligand-cell surface receptor interaction. This interaction results in activation of
receptor-associated tyrosine kinases, which subsequently activate other cytosolic targets, which
then initiate various signaling pathways. The major pathways regulated by celastrol are described
in the text and shown in the figure. Celastrol is known to target one or more of these pathways
leading to the suppression of inflammation associated with various chronic diseases including,
inflammatory diseases, autoimmune diseases, obesity, and cancer. There is no direct evidence for
celastrol-induced modulation of calmodulin pathway. However, regulation of calcium release and
AMPK activity by celastrol suggests the possibility of regulation of the calmodulin pathway by
celsatrol. Besides cell signaling, celastrol modulates other disease-related processes (e.g.,
angiogenesis, heat-shock protein responses, and proteasome activity) to limit disease progression
and to facilitate recovery, where feasible.
Therefore, additional studies are needed to clearly understand the role of celastrol in
the regulation of mTOR and HIF-1.
Celastrol possesses antioxidant activity. Oxidative stress is one of the mediators
of inflammation [73]. Oxidative stress builds up with the generation of high levels
of reactive free radicals, such as reactive oxygen species (ROS; e.g., chemically
reactive molecules derived from O2 mainly O2− (superoxide anions),
H2O2 (hydrogen peroxide) and •OH (hydroxyl radicals)) and reactive nitrogen
species (RNS; e.g., radicals derived from nitrogen and oxygen, particularly nitric
oxide (NO)) in the cell [74, 75]. Celastrol is shown to inhibit lipid peroxidation in
rat liver mitochondria by direct radical scavenging [76] as well as neutralizing
oxygen radicals [77]. Celastrol also enhanced the antioxidant defense system and
offered protection against bleomycin-induced pulmonary fibrosis in rats by restor-
ing antioxidant enzymes such as hemoxygenase-1 (HO-1),
glutathione-S-transferase (GSTs) and nicotinamide adenine dinucleotide phosphate
(H) (NADP(H)): quinine oxidoreductase (NQO1) via the NF-E2-related factor-2
(Nrf2) pathway [78]. Similarly, celastrol decreased obesity-induced oxidative stress
by increasing antioxidant enzymes and inhibiting NADH oxidase and ROS [79].
Defense against oxidant system by celastrol has also been attributed to decreased
expression of iNOS and NO production [30, 34], and the blocking of reactive thiols
[17]. In contrast to the above, celastrol has been reported to induce ROS accu-
mulation and to initiate apoptosis through the downregulation on Hsp90 in tumor
cells [80]. Similarly, in osteosarcoma, celastrol caused G2/M phase arrest, and
induced apoptosis and autophagy via the ROS/JNK signaling pathway [81].
Celastrol is known to have anticancer and anti-metastatic activities [15, 82, 83, 107,
108]. In addition, celastrol is shown to enhance the therapeutic efficacy of other
anticancer drugs when used with them, and to potentiate the beneficial effects of
radiotherapy [109, 110]. The major processes involved in these activities and
affected by celastrol include, the inhibition of cellular proliferation, induction of
apoptosis, prevention of malignant tissue invasion, and blockade of angiogenesis
[7, 15, 82, 111]. Celastrol can inhibit cell proliferation and induce apoptosis via
multiple actions. These include, potentiation of TNF-induced apoptosis via sup-
pression of the NF-kB pathway [4]; downregulation of cytokines such as IL-6,
which is an inducer of cell proliferation [112]; activation of caspases [113–115];
inhibition of the expression of anti-apoptotic proteins such as cellular inhibitor of
apoptosis protein 1 and 2 (cIAP1 and cIAP2), cellular FLICE-inhibitory protein
(FLIP), and B-cell lymphoma 2 (Bcl-2) [4, 114]; induction of cell cycle arrest [81,
116]; and downregulation of cell survival proteins coupled with upregulation of
death receptors [117]. Furthermore, celastrol inhibits adhesion, migration and
invasion of tumor cells via reduced expression of specific integrins, as well as
reduced MMP activity [118–120]. In addition, as described above, celastrol can
suppress angiogenesis [63, 83].
Two additional mechanisms contribute to the anticancer effects of celastrol,
namely inhibition of Hsp90 and proteasome inhibition. In regard to Hsp90, celastrol
directly binds to the C-terminal domain of Hsp90 inducing its oligomerization, and
it interferes with specific biological functions through modulation of
Hsp90-associated nuclear transcription factors [106, 121]. In addition, celastrol has
12 Celastrol and Its Role in Controlling Chronic Diseases 279
the treatment of RA, SLE, and other disorders [1–3, 5, 127]. However, a large part
of this information is based on folklores as well as documented description of the
use of these herbal products in old archived literature. Limited available information
is derived from studies on small numbers of patients and/or scientifically controlled
randomized clinical studies on the use of T. wilfordii in chronic inflammatory and
autoimmune diseases such as RA, juvenile RA, ankylosing spondylitis, and SLE [5,
128–132]. Of these, the most reliable clinical studies have been performed using T.
wilfordii in patients with RA. The efficacy of T. wilfordii extract against RA was
compared with that of two of the mainstream anti-arthritic drugs, namely sul-
phasalazine and methotrexate. Interestingly, T. wilfordii extract reduced the severity
of RA as assessed by well-established criteria, and the efficacy of T. wilfordii was
comparable to, or better than, that of sulphasalazine/ methotrexate [128, 133–136].
Furthermore, the combination of T. wilfordii and methotrexate was better than
methotrexate alone. However, the toxicity profile of this natural product needs
further assessment before it can be approved for therapeutic purposes.
12.9 Conclusions
Acknowledgments We thank Dr. Hua Yu, Dr. Brian Astry, Dr. Siddaraju Nanjundaiah, Dr.
Rajesh Rajaiah, and Dr. Li Tong for their contribution to the original studies based on celastrol as
well as for their helpful discussions.
Funding This work was supported by National Institutes of Health (NIH)/National Center for
Complementary and Integrative Health (NCCIH) Grant Number AT004321.
12 Celastrol and Its Role in Controlling Chronic Diseases 281
References
1. Jin HZ, Hwang BY, Kim HS, Lee JH, Kim YH, Lee JJ (2002) Antiinflammatory constituents
of Celastrus orbiculatus inhibit the NF-kappaB activation and NO production. J Nat Prod 65
(1):89–91
2. Luo DQ, Wang H, Tian X, Shao HJ, Liu JK (2005) Antifungal properties of pristimerin and
celastrol isolated from Celastrus hypoleucus. Pest Manag Sci 61(1):85–90. doi:10.1002/ps.
953
3. Tong L, Moudgil KD (2007) Celastrus aculeatus Merr. suppresses the induction and
progression of autoimmune arthritis by modulating immune response to heat-shock protein
65. Arthritis Res Ther 9(4):R70. doi:10.1186/ar2268
4. Sethi G, Ahn KS, Pandey MK, Aggarwal BB (2007) Celastrol, a novel triterpene, potentiates
TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting
NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation. Blood
109(7):2727–2735. doi:10.1182/blood-2006-10-050807
5. Wong KF, Yuan Y, Luk JM (2012) Tripterygium wilfordii bioactive compounds as
anticancer and anti-inflammatory agents. Clin Exp Pharmacol Physiol 39(3):311–320.
doi:10.1111/j.1440-1681.2011.05586.x
6. Salminen A, Lehtonen M, Paimela T, Kaarniranta K (2010) Celastrol: molecular targets of
thunder god vine. Biochem Biophys Res Commun 394(3):439–442. doi:10.1016/j.bbrc.2010.
03.050
7. Kannaiyan R, Shanmugam MK, Sethi G (2011) Molecular targets of celastrol derived from
Thunder of God Vine: potential role in the treatment of inflammatory disorders and cancer.
Cancer Lett 303(1):9–20. doi:10.1016/j.canlet.2010.10.025
8. Venkatesha SH, Yu H, Rajaiah R, Tong L, Moudgil KD (2011) Celastrus-derived celastrol
suppresses autoimmune arthritis by modulating antigen-induced cellular and humoral effector
responses. J Biol Chem 286(17):15138–15146. doi:10.1074/jbc.M111.226365
9. Nanjundaiah SM, Venkatesha SH, Yu H, Tong L, Stains JP, Moudgil KD (2012) Celastrus
and its bioactive celastrol protect against bone damage in autoimmune arthritis by
modulating osteoimmune cross-talk. J Biol Chem 287(26):22216–22226. doi:10.1074/jbc.
M112.356816
10. Wang Y, Cao L, Xu LM, Cao FF, Peng B, Zhang X, Shen YF, Uzan G, Zhang DH (2015)
Celastrol ameliorates EAE induction by suppressing pathogenic T cell responses in the
peripheral and central nervous systems. J Neuroimmune Pharmacol 10(3):506–516. doi:10.
1007/s11481-015-9598-9
11. Shaker ME, Ashamallah SA, Houssen ME (2014) Celastrol ameliorates murine colitis via
modulating oxidative stress, inflammatory cytokines and intestinal homeostasis. Chem Biol
Interact 210:26–33. doi:10.1016/j.cbi.2013.12.007
12. Faust K, Gehrke S, Yang Y, Yang L, Beal MF, Lu B (2009) Neuroprotective effects of
compounds with antioxidant and anti-inflammatory properties in a Drosophila model of
Parkinson’s disease. BMC Neurosci 10:109. doi:10.1186/1471-2202-10-109
13. Liu Z, Ma L, Zhou GB (2011) The main anticancer bullets of the Chinese medicinal herb,
thunder god vine. Molecules 16(6):5283–5297. doi:10.3390/molecules16065283
14. Abbas Bukhari SN, Jantan I, Seyed MA (2015) Effects of plants and isolates of celastraceae
family on cancer pathways. Anticancer Agents Med Chem 15(6):681–693
15. Yadav VR, Prasad S, Sung B, Kannappan R, Aggarwal BB (2010) Targeting inflammatory
pathways by triterpenoids for prevention and treatment of cancer. Toxins (Basel) 2
(10):2428–2466. doi:10.3390/toxins2102428
16. Lee JH, Koo TH, Yoon H, Jung HS, Jin HZ, Lee K, Hong YS, Lee JJ (2006) Inhibition of
NF-kappa B activation through targeting I kappa B kinase by celastrol, a quinone methide
triterpenoid. Biochem Pharmacol 72(10):1311–1321. doi:10.1016/j.bcp.2006.08.014
17. Trott A, West JD, Klaic L, Westerheide SD, Silverman RB, Morimoto RI, Morano KA
(2008) Activation of heat shock and antioxidant responses by the natural product celastrol:
282 S.H. Venkatesha and K.D. Moudgil
35. Kiaei M, Kipiani K, Petri S, Chen J, Calingasan NY, Beal MF (2005) Celastrol blocks
neuronal cell death and extends life in transgenic mouse model of amyotrophic lateral
sclerosis. Neurodegener Dis 2(5):246–254. doi:10.1159/000090364
36. Paris D, Ganey NJ, Laporte V, Patel NS, Beaulieu-Abdelahad D, Bachmeier C, March A,
Ait-Ghezala G, Mullan MJ (2010) Reduction of beta-amyloid pathology by celastrol in a
transgenic mouse model of Alzheimer’s disease. J Neuroinflammation 7:17. doi:10.1186/
1742-2094-7-17
37. Tabuchi H, Konishi M, Saito N, Kato M, Mimura M (2014) Reverse Fox test for detecting
visuospatial dysfunction corresponding to parietal hypoperfusion in mild Alzheimer’s
disease. Am J Alzheimers Dis Other Demen 29(2):177–182. doi:10.1177/
1533317513511291
38. Yang C, Swallows CL, Zhang C, Lu J, Xiao H, Brady RO, Zhuang Z (2014) Celastrol
increases glucocerebrosidase activity in Gaucher disease by modulating molecular
chaperones. Proc Natl Acad Sci USA 111(1):249–254. doi:10.1073/pnas.1321341111
39. Paimela T, Hyttinen JM, Viiri J, Ryhanen T, Karjalainen RO, Salminen A, Kaarniranta K
(2011) Celastrol regulates innate immunity response via NF-kappaB and Hsp70 in human
retinal pigment epithelial cells. Pharmacol Res 64(5):501–508. doi:10.1016/j.phrs.2011.05.
027
40. Hu H, Straub A, Tian Z, Bassler N, Cheng J, Peter K (2009) Celastrol, a triterpene extracted
from Tripterygium wilfordii Hook F, inhibits platelet activation. J Cardiovasc Pharmacol 54
(3):240–245. doi:10.1097/FJC.0b013e3181b21472
41. Gu L, Bai W, Li S, Zhang Y, Han Y, Gu Y, Meng G, Xie L, Wang J, Xiao Y, Shan L,
Zhou S, Wei L, Ferro A, Ji Y (2013) Celastrol prevents atherosclerosis via inhibiting LOX-1
and oxidative stress. PLoS ONE 8(6):e65477
42. Liu J, Lee J, Salazar Hernandez MA, Mazitschek R, Ozcan U (2015) Treatment of obesity
with celastrol. Cell 161(5):999–1011. doi:10.1016/j.cell.2015.05.011
43. Kim JE, Lee MH, Nam DH, Song HK, Kang YS, Lee JE, Kim HW, Cha JJ, Hyun YY,
Han SY, Han KH, Han JY, Cha DR (2013) Celastrol, an NF-kappaB inhibitor, improves
insulin resistance and attenuates renal injury in db/db mice. PLoS ONE 8(4):e62068
44. Youn GS, Kwon DJ, Ju SM, Rhim H, Bae YS, Choi SY, Park J (2014) Celastrol ameliorates
HIV-1 Tat-induced inflammatory responses via NF-kappaB and AP-1 inhibition and heme
oxygenase-1 induction in astrocytes. Toxicol Appl Pharmacol 280(1):42–52. doi:10.1016/j.
taap.2014.07.010
45. Tallorin L, Durrant JD, Nguyen QG, McCammon JA, Burkart MD (2014) Celastrol inhibits
Plasmodium falciparum enoyl-acyl carrier protein reductase. Bioorganic Med Chem 22
(21):6053–6061
46. Simmonds RE, Foxwell BM (2008) Signalling, inflammation and arthritis: NF-kappaB and
its relevance to arthritis and inflammation. Rheumatology (Oxford) 47(5):584–590. doi:10.
1093/rheumatology/kem298
47. Tak PP, Firestein GS (2001) NF-kappaB: a key role in inflammatory diseases. J Clin Invest
107(1):7–11. doi:10.1172/JCI11830
48. Bondeson J, Foxwell B, Brennan F, Feldmann M (1999) Defining therapeutic targets by
using adenovirus: blocking NF-kappaB inhibits both inflammatory and destructive
mechanisms in rheumatoid synovium but spares anti-inflammatory mediators. Proc Natl
Acad Sci USA 96(10):5668–5673
49. Ju SM, Youn GS, Cho YS, Choi SY, Park J (2015) Celastrol ameliorates cytokine toxicity
and pro-inflammatory immune responses by suppressing NF-kappaB activation in RINm5F
beta cells. BMB Rep 48(3):172–177
50. Hommes DW, Peppelenbosch MP, van Deventer SJ (2003) Mitogen activated protein
(MAP) kinase signal transduction pathways and novel anti-inflammatory targets. Gut 52
(1):144–151
51. Kaminska B (2005) MAPK signalling pathways as molecular targets for anti-inflammatory
therapy–from molecular mechanisms to therapeutic benefits. Biochim Biophys Acta 1754(1–
2):253–262. doi:10.1016/j.bbapap.2005.08.017
284 S.H. Venkatesha and K.D. Moudgil
52. Tan Y, Rouse J, Zhang A, Cariati S, Cohen P, Comb MJ (1996) FGF and stress regulate
CREB and ATF-1 via a pathway involving p38 MAP kinase and MAPKAP kinase-2.
EMBO J 15(17):4629–4642
53. Pierrat B, Correia JS, Mary JL, Tomas-Zuber M, Lesslauer W (1998) RSK-B, a novel
ribosomal S6 kinase family member, is a CREB kinase under dominant control of p38alpha
mitogen-activated protein kinase (p38alphaMAPK). J Biol Chem 273(45):29661–29671
54. Jung HW, Chung YS, Kim YS, Park YK (2007) Celastrol inhibits production of nitric oxide
and proinflammatory cytokines through MAPK signal transduction and NF-kappaB in
LPS-stimulated BV-2 microglial cells. Exp Mol Med 39(6):715–721. doi:10.1038/emm.
2007.78
55. Kannaiyan R, Manu KA, Chen L, Li F, Rajendran P, Subramaniam A, Lam P, Kumar AP,
Sethi G (2011) Celastrol inhibits tumor cell proliferation and promotes apoptosis through the
activation of c-Jun N-terminal kinase and suppression of PI3 K/Akt signaling pathways.
Apoptosis 16(10):1028–1041
56. Kim DH, Shin EK, Kim YH, Lee BW, Jun JG, Park JH, Kim JK (2009) Suppression of
inflammatory responses by celastrol, a quinone methide triterpenoid isolated from Celastrus
regelii. Eur J Clin Investig 39(9):819–827. doi:10.1111/j.1365-2362.2009.02186.x
57. Zhu H, Liu XW, Cai TY, Cao J, Tu CX, Lu W, He QJ, Yang B (2010) Celastrol acts as a
potent antimetastatic agent targeting beta1 integrin and inhibiting cell-extracellular matrix
adhesion, in part via the p38 mitogen-activated protein kinase pathway. J Pharmacol Exp
Ther 334(2):489–499. doi:10.1124/jpet.110.165654
58. Shuai K, Liu B (2003) Regulation of JAK-STAT signalling in the immune system. Nat Rev
Immunol 3(11):900–911. doi:10.1038/nri1226
59. Darnell JE Jr (1997) STATs and gene regulation. Science 277(5332):1630–1635
60. Kannaiyan R, Hay HS, Rajendran P, Li F, Shanmugam MK, Vali S, Abbasi T, Kapoor S,
Sharma A, Kumar AP, Chng WJ, Sethi G (2011) Celastrol inhibits proliferation and induces
chemosensitization through down-regulation of NF-kappaB and STAT3 regulated gene
products in multiple myeloma cells. Br J Pharmacol 164(5):1506–1521. doi:10.1111/j.1476-
5381.2011.01449.x
61. Rajendran P, Li F, Shanmugam MK, Kannaiyan R, Goh JN, Wong KF, Wang W, Khin E,
Tergaonkar V, Kumar AP, Luk JM, Sethi G (2012) Celastrol suppresses growth and induces
apoptosis of human hepatocellular carcinoma through the modulation of STAT3/JAK2
signaling cascade in vitro and in vivo. Cancer Prev Res (Phila) 5(4):631–643. doi:10.1158/
1940-6207.CAPR-11-0420
62. Chen S, Gu C, Xu C, Zhang J, Xu Y, Ren Q, Guo M, Huang S, Chen L (2014) Celastrol
prevents cadmium-induced neuronal cell death via targeting JNK and PTEN-Akt/mTOR
network. J Neurochem 128(2):256–266. doi:10.1111/jnc.12474
63. Ma J, Han LZ, Liang H, Mi C, Shi H, Lee JJ, Jin X (2014) Celastrol inhibits the HIF-1alpha
pathway by inhibition of mTOR/p70S6 K/eIF4E and ERK1/2 phosphorylation in human
hepatoma cells. Oncol Rep 32(1):235–242. doi:10.3892/or.2014.3211
64. Mabuchi S, Kuroda H, Takahashi R, Sasano T (2015) The PI3 K/AKT/mTOR pathway as a
therapeutic target in ovarian cancer. Gynecol Oncol 137(1):173–179. doi:10.1016/j.ygyno.
2015.02.003
65. Sha M, Ye J, Zhang LX, Luan ZY, Chen YB, Huang JX (2014) Celastrol induces apoptosis
of gastric cancer cells by miR-21 inhibiting PI3 K/Akt-NF-kappaB signaling pathway.
Pharmacology 93(1–2):39–46. doi:10.1159/000357683
66. Shrivastava S, Jeengar MK, Reddy VS, Reddy GB, Naidu VG (2015) Anticancer effect of
celastrol on human triple negative breast cancer: possible involvement of oxidative stress,
mitochondrial dysfunction, apoptosis and PI3 K/Akt pathways. Exp Mol Pathol 98(3):313–
327. doi:10.1016/j.yexmp.2015.03.031
67. Zhao J, Sun Y, Shi P, Dong JN, Zuo LG, Wang HG, Gong JF, Li Y, Gu LL, Li N, Li JS,
Zhu WM (2015) Celastrol ameliorates experimental colitis in IL-10 deficient mice via the
up-regulation of autophagy. Int Immunopharmacol 26(1):221–228. doi:10.1016/j.intimp.
2015.03.033
12 Celastrol and Its Role in Controlling Chronic Diseases 285
68. Polivka J Jr, Janku F (2014) Molecular targets for cancer therapy in the PI3 K/AKT/mTOR
pathway. Pharmacol Ther 142(2):164–175. doi:10.1016/j.pharmthera.2013.12.004
69. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC, Kaper F, Giaccia AJ,
Abraham RT (2002) Regulation of hypoxia-inducible factor 1alpha expression and function
by the mammalian target of rapamycin. Mol Cell Biol 22(20):7004–7014
70. Han X, Sun S, Zhao M, Cheng X, Chen G, Lin S, Guan Y, Yu X (2014) Celastrol stimulates
hypoxia-inducible factor-1 activity in tumor cells by initiating the ROS/Akt/p70S6 K
signaling pathway and enhancing hypoxia-inducible factor-1alpha protein synthesis.
PLoS ONE 9(11):e112470. doi:10.1371/journal.pone.0112470
71. Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouyssegur J, Mazure NM (2009)
Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of
BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol 29(10):2570–2581. doi:10.1128/
MCB.00166-09
72. Onnis B, Rapisarda A, Melillo G (2009) Development of HIF-1 inhibitors for cancer therapy.
J Cell Mol Med 13(9A):2780–2786. doi:10.1111/j.1582-4934.2009.00876.x
73. Holmstrom KM, Finkel T (2014) Cellular mechanisms and physiological consequences of
redox-dependent signalling. Nat Rev Mol Cell Biol 15(6):411–421. doi:10.1038/nrm3801
74. Ye ZW, Zhang J, Townsend DM, Tew KD (2015) Oxidative stress, redox regulation and
diseases of cellular differentiation. Biochim Biophys Acta 1850(8):1607–1621. doi:10.1016/
j.bbagen.2014.11.010
75. Halliwell B (1987) Free radicals and metal ions in health and disease. Proc Nutr Soc 46
(1):13–26
76. Sassa H, Takaishi Y, Terada H (1990) The triterpene celastrol as a very potent inhibitor of
lipid peroxidation in mitochondria. Biochem Biophys Res Commun 172(2):890–897
77. Sassa H, Kogure K, Takaishi Y, Terada H (1994) Structural basis of potent antiperoxidation
activity of the triterpene celastrol in mitochondria: effect of negative membrane surface
charge on lipid peroxidation. Free Radic Biol Med 17(3):201–207
78. Divya T, Dineshbabu V, Soumyakrisnan S, Sureshkumar S, Sudandiran G (2016) Celastrol
enhances Nrf2 mediated antioxidant enzymes and exhibits anti-fibrotic effect through
regulation of collagen production against bleomycin-induced pulmonary fibrosis. Chem Biol
Interact 246:52–62
79. Wang C, Shi C, Yang X, Yang M, Sun H, Wang C (2014) Celastrol suppresses obesity
process via increasing antioxidant capacity and improving lipid metabolism. Eur J Pharmacol
744:52–58. doi:10.1016/j.ejphar.2014.09.043
80. Chen G, Zhang X, Zhao M, Wang Y, Cheng X, Wang D, Xu Y, Du Z, Yu X (2011) Celastrol
targets mitochondrial respiratory chain complex I to induce reactive oxygen
species-dependent cytotoxicity in tumor cells. BMC Cancer 11:170. doi:10.1186/1471-
2407-11-170
81. Li HY, Zhang J, Sun LL, Li BH, Gao HL, Xie T, Zhang N, Ye ZM (2015) Celastrol induces
apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells:
an in vitro and in vivo study. Cell Death Dis 6:e1604. doi:10.1038/cddis.2014.543
82. Gupta SC, Kim JH, Prasad S, Aggarwal BB (2010) Regulation of survival, proliferation,
invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory
pathways by nutraceuticals. Cancer Metastas Rev 29(3):405–434. doi:10.1007/s10555-010-
9235-2
83. Pang X, Yi Z, Zhang J, Lu B, Sung B, Qu W, Aggarwal BB, Liu M (2010) Celastrol
suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian
target of rapamycin pathway. Cancer Res 70(5):1951–1959
84. Khan KA, Bicknell R (2015) Anti-angiogenic alternatives to VEGF blockade. Clin Exp
Metastas 33(2):197–210
85. Szekanecz Z, Besenyei T, Paragh G, Koch AE (2009) Angiogenesis in rheumatoid arthritis.
Autoimmunity 42(7):563–573
86. Koch AE (2000) The role of angiogenesis in rheumatoid arthritis: recent developments. Ann
Rheum Dis 59(Suppl 1):i65–i71
286 S.H. Venkatesha and K.D. Moudgil
106. Zhang D, Xu L, Cao F, Wei T, Yang C, Uzan G, Peng B (2010) Celastrol regulates multiple
nuclear transcription factors belonging to HSP90’s clients in a dose- and cell type-dependent
way. Cell Stress Chaperones 15(6):939–946
107. Petronelli A, Pannitteri G, Testa U (2009) Triterpenoids as new promising anticancer drugs.
Anticancer Drugs 20(10):880–892. doi:10.1097/CAD.0b013e328330fd90
108. Yadav VR, Sung B, Prasad S, Kannappan R, Cho SG, Liu M, Chaturvedi MM, Aggarwal BB
(2010) Celastrol suppresses invasion of colon and pancreatic cancer cells through the
downregulation of expression of CXCR4 chemokine receptor. J Mol Med 88(12):1243–1253
109. Zheng L, Fu Y, Zhuang L, Gai R, Ma J, Lou J, Zhu H, He Q, Yang B (2014) Simultaneous
NF-kappaB inhibition and E-cadherin upregulation mediate mutually synergistic anticancer
activity of celastrol and SAHA in vitro and in vivo. Int J Cancer 135(7):1721–1732. doi:10.
1002/ijc.28810
110. Dai Y, DeSano JT, Meng Y, Ji Q, Ljungman M, Lawrence TS, Xu L (2009) Celastrol
potentiates radiotherapy by impairment of DNA damage processing in human prostate
cancer. Int J Radiat Oncol Biol Phys 74(4):1217–1225. doi:10.1016/j.ijrobp.2009.03.057
111. Li-Weber M (2013) Targeting apoptosis pathways in cancer by Chinese medicine. Cancer
Lett 332(2):304–312. doi:10.1016/j.canlet.2010.07.015
112. Chiang KC, Tsui KH, Chung LC, Yeh CN, Chen WT, Chang PL, Juang HH (2014) Celastrol
blocks interleukin-6 gene expression via downregulation of NF-kappaB in prostate
carcinoma cells. PLoS ONE 9(3):e93151. doi:10.1371/journal.pone.0093151
113. Lu L, Shi W, Deshmukh RR, Long J, Cheng X, Ji W, Zeng G, Chen X, Zhang Y, Dou QP
(2014) Tumor necrosis factor-alpha sensitizes breast cancer cells to natural products with
proteasome-inhibitory activity leading to apoptosis. PLoS ONE 9(11):e113783. doi:10.1371/
journal.pone.0113783
114. Mi C, Shi H, Ma J, Han LZ, Lee JJ, Jin X (2014) Celastrol induces the apoptosis of breast
cancer cells and inhibits their invasion via downregulation of MMP-9. Oncol Rep 32
(6):2527–2532. doi:10.3892/or.2014.3535
115. Yang HS, Kim JY, Lee JH, Lee BW, Park KH, Shim KH, Lee MK, Seo KI (2011) Celastrol
isolated from Tripterygium regelii induces apoptosis through both caspase-dependent and -
independent pathways in human breast cancer cells. Food Chem Toxicol 49(2):527–532.
doi:10.1016/j.fct.2010.11.044
116. Peng B, Xu L, Cao F, Wei T, Yang C, Uzan G, Zhang D (2010) HSP90 inhibitor, celastrol,
arrests human monocytic leukemia cell U937 at G0/G1 in thiol-containing agents reversible
way. Mol Cancer 9:79. doi:10.1186/1476-4598-9-79
117. Sung B, Park B, Yadav VR, Aggarwal BB (2010) Celastrol, a triterpene, enhances
TRAIL-induced apoptosis through the down-regulation of cell survival proteins and
up-regulation of death receptors. J Biol Chem 285(15):11498–11507
118. Li GQ, Liu D, Zhang Y, Qian YY, Zhu YD, Guo SY, Sunagawa M, Hisamitsu T, Liu YQ
(2013) Anti-invasive effects of celastrol in hypoxia-induced fibroblast-like synoviocyte
through suppressing of HIF-1alpha/CXCR4 signaling pathway. Int Immunopharmacol 17
(4):1028–1036. doi:10.1016/j.intimp.2013.10.006
119. Xu J, Wu CL, Huang J (2015) Effect of celastrol in inhibiting metastasis of lung cancer cells
by influencing Akt signaling pathway and expressing integrins. Zhongguo Zhong Yao Za Zhi
40(6):1129–1133
120. Xu J, Wu CL (2015) Anti-metastasis of celastrol on esophageal cancer cells and its
mechanism. Sheng Li Xue Bao 67(3):341–347
121. Zanphorlin LM, Alves FR, Ramos CH (2014) The effect of celastrol, a triterpene with
antitumorigenic activity, on conformational and functional aspects of the human 90 kDa heat
shock protein Hsp90alpha, a chaperone implicated in the stabilization of the tumor
phenotype. Biochim Biophys Acta 10:3145–3152. doi:10.1016/j.bbagen.2014.06.008
122. Wang WB, Feng LX, Yue QX, Wu WY, Guan SH, Jiang BH, Yang M, Liu X, Guo DA
(2012) Paraptosis accompanied by autophagy and apoptosis was induced by celastrol, a
natural compound with influence on proteasome, ER stress and Hsp90. J Cell Physiol 227
(5):2196–2206
288 S.H. Venkatesha and K.D. Moudgil
123. Yang H, Landis-Piwowar KR, Chen D, Milacic V, Dou QP (2008) Natural compounds with
proteasome inhibitory activity for cancer prevention and treatment. Curr Protein Pept Sci 9
(3):227–239
124. Dou QP, Zonder JA (2014) Overview of proteasome inhibitor-based anticancer therapies:
perspective on bortezomib and second generation proteasome inhibitors versus future
generation inhibitors of ubiquitin-proteasome system. Curr Cancer Drug Targets 14(6):517–
536
125. Hoesel B, Schmid JA (2013) The complexity of NF-kappaB signaling in inflammation and
cancer. Mol Cancer 12:86
126. Idris AI, Libouban H, Nyangoga H, Landao-Bassonga E, Chappard D, Ralston SH (2009)
Pharmacologic inhibitors of IkappaB kinase suppress growth and migration of mammary
carcinosarcoma cells in vitro and prevent osteolytic bone metastasis in vivo. Mol Cancer
Ther 8(8):2339–2347. doi:10.1158/1535-7163.MCT-09-0133
127. Wang KW, Mao JS, Tai YP, Pan YJ (2006) Novel skeleton terpenes from Celastrus
hypoleucus with anti-tumor activities. Bioorg Med Chem Lett 16(8):2274–2277. doi:10.
1016/j.bmcl.2006.01.021
128. Tao X, Cush JJ, Garret M, Lipsky PE (2001) A phase I study of ethyl acetate extract of the
chinese antirheumatic herb Tripterygium wilfordii hook F in rheumatoid arthritis.
J Rheumatol 28(10):2160–2167
129. Zhang W, Shi Q, Zhao LD, Li Y, Tang FL, Zhang FC, Zhang X (2010) The safety and
effectiveness of a chloroform/methanol extract of Tripterygium wilfordii Hook F (T2) plus
methotrexate in treating rheumatoid arthritis. J Clin Rheumatol 16(8):375–378. doi:10.1097/
RHU.0b013e3181fe8ad1
130. Ji W, Li J, Lin Y, Song YN, Zhang M, Ke Y, Ren Y, Deng X, Zhang J, Huang F, Yu D
(2010) Report of 12 cases of ankylosing spondylitis patients treated with Tripterygium
wilfordii. Clin Rheumatol 29(9):1067–1072. doi:10.1007/s10067-010-1497-0
131. Gao ZG, Zang AC, Bai RX (1986) Radix Tripterygium Wilfordii Hook F in rheumatoid
arthritis, ankylosing spondylitis and juvenile rheumatoid arthritis. Chin Med J (Engl) 99
(4):317–320
132. Patavino T, Brady DM (2001) Natural medicine and nutritional therapy as an alternative
treatment in systemic lupus erythematosus. Altern Med Rev 6(5):460–471
133. Tao X, Younger J, Fan FZ, Wang B, Lipsky PE (2002) Benefit of an extract of Tripterygium
Wilfordii Hook F in patients with rheumatoid arthritis: a double-blind, placebo-controlled
study. Arthritis Rheum 46(7):1735–1743
134. Goldbach-Mansky R, Wilson M, Fleischmann R, Olsen N, Silverfield J, Kempf P, Kivitz A,
Sherrer Y, Pucino F, Csako G, Costello R, Pham TH, Snyder C, van der Heijde D, Tao X,
Wesley R, Lipsky PE (2009) Comparison of Tripterygium wilfordii Hook F versus
sulfasalazine in the treatment of rheumatoid arthritis: a randomized trial. Ann Intern Med 151
(4):229–240
135. Lv QW, Zhang W, Shi Q, Zheng WJ, Li X, Chen H, Wu QJ, Jiang WL, Li HB, Gong L,
Wei W, Liu H, Liu AJ, Jin HT, Wang JX, Liu XM, Li ZB, Liu B, Shen M, Wang Q, Wu XN,
Liang D, Yin YF, Fei YY, Su JM, Zhao LD, Jiang Y, Li J, Tang FL, Zhang FC, Lipsky PE,
Zhang X (2014) Comparison of Tripterygium wilfordii Hook F with methotrexate in the
treatment of active rheumatoid arthritis (TRIFRA): a randomised, controlled clinical trial.
Ann Rheum Dis. doi:10.1136/annrheumdis-2013-204807
136. Moudgil KD, Berman BM (2014) Traditional Chinese medicine: potential for clinical
treatment of rheumatoid arthritis. Expert Rev Clin Immunol 10(7):819–822
137. Li H, Zhang YY, Tan HW, Jia YF, Li D (2008) Therapeutic effect of tripterine on adjuvant
arthritis in rats. J Ethnopharmacol 118(3):479–484. doi:10.1016/j.jep.2008.05.028
138. Li H, Jia YF, Pan Y, Pan DJ, Li D, Zhang LX (1997) Effect of tripterine on collagen-induced
arthritis in rats. Zhongguo Yao Li Xue Bao 18(3):270–273
139. Xu Z, Wu G, Wei X, Chen X, Wang Y, Chen L (2013) Celastrol induced DNA damage, cell
cycle arrest, and apoptosis in human rheumatoid fibroblast-like synovial cells. Am J Chin
Med 41(3):615–628. doi:10.1142/S0192415X13500432
12 Celastrol and Its Role in Controlling Chronic Diseases 289
140. Li GQ, Zhang Y, Liu D, Qian YY, Zhang H, Guo SY, Sunagawa M, Hisamitsu T, Liu YQ
(2012) Celastrol inhibits interleukin-17A-stimulated rheumatoid fibroblast-like synoviocyte
migration and invasion through suppression of NF-kappaB-mediated matrix
metalloproteinase-9 expression. Int Immunopharmacol 14(4):422–431. doi:10.1016/j.
intimp.2012.08.016
141. Zhou LL, Lin ZX, Fung KP, Cheng CH, Che CT, Zhao M, Wu SH, Zuo Z (2011)
Celastrol-induced apoptosis in human HaCaT keratinocytes involves the inhibition of
NF-kappaB activity. Eur J Pharmacol 670(2–3):399–408. doi:10.1016/j.ejphar.2011.09.014
142. Zhu F, Li C, Jin XP, Weng SX, Fan LL, Zheng Z, Li WL, Wang F, Wang WF, Hu XF,
Lv CL, Liu P (2014) Celastrol may have an anti-atherosclerosis effect in a rabbit
experimental carotid atherosclerosis model. Int J Clin Exp Med 7(7):1684–1691
143. Venkatesha SH, Dudics S, Astry B, Moudgil KD (2016) Control of autoimmune
inflammation by celastrol, a natural triterpenoid. Pathog Dis 74(6). pii:ftw059. doi: 10.
1093/femspd/ftw059
Chapter 13
Boswellic Acids and Their Role in Chronic
Inflammatory Diseases
H.P.T. Ammon
Abstract Boswellic acids, which are pentacyclic triterpenes belong to the active
pharmacological compounds of the oleogum resin of different Boswellia species. In
the resin, more than 12 different boswellic acids have been identified but only KBA
and AKBA received significant pharmacological interest. Biological Activity: In an
extract of the resin of Boswellia species multiple factors are responsible for the final
outcome of a therapeutic effect, be it synergistic or antagonistic. Moreover, the
anti-inflammatory actions of BAs are caused by different mechanisms of action.
They include inhibition of leukotriene synthesis and to a less extend prostaglandin
synthesis. Furthermore inhibition of the complement system at the level of con-
version of C3 into C3a and C3b. A major target of BAs is the immune system. Here,
BEs as well as BAs including KBA and AKBA, have been shown to decrease
production of proinflammatory cytokines including IL-1, IL-2, IL-6, IFN-c and
TNF-a which finally are directed to destroy tissues such as cartilage, insulin pro-
ducing cells, bronchial, intestinal and other tissues. NFĸB is considered to be the
target of AKBA. The complex actions of BEs and BAs in inflamed areas may be
completed by some effects that are localized behind the inflammatory process as
such tissue destruction. In this case, in vitro- and animal studies have shown that
BAs and BEs suppress proteolytic activity of cathepsin G, human leucocyte elas-
tase, formation of oxygen radicals and lysosomal enzymes. Pharmacokinetics:
Whereas KBA is absorbed reaching blood levels being close to in vitro IC50, AKBA
which is more active in in vitro studies than KBA, but undergoes much less
absorption than KBA. However, absorption of both is increased more than twice
when taken together with a high-fat meal.Clinical Studies There are a variety of
chronic inflammatory diseases which respond to treatment with extracts from the
resin of Boswellia species. Though, the number of cases is small in related clinical
studies, their results are convincing and supported by the preclinical data. These
Keywords Boswellic acids Boswellic extracts Leukotrienes Prostaglandins
Proteolytic enzymes Cytokines Pharmacokinetics Rheumatoid arthritis
Inflammatory bowel diseases Bronchial asthma Diabetes Side effects
Abbreviations
AA Arachidonic Acid
ABA Acetyl-boswellic acid
Ac-OH-LA 3a-Acetoxy-28-hydroxylup-20 (29)
AKBA Acetyl-11-keto-b-Boswellic acid
a-BA a-Boswellic acid
b-BA b-Boswellic acid
BA Boswellic acid
BE Boswellic extract
BS Boswellia serrata
BSA Bovine serum albumin
CD4 CD4 lymphocytes
CD8 CD8 lymphocytes
CIA Collagen-induced arthritis
COX Cyclooxygenase
CRP C-reactive protein
CDAI Crohn’s Disease Activity Index
ConA Concavalin A
ESR Erythrocyte Sedimentation Rate
FCV Forced vital capacity
FEV1 Forced expiratory volume
GSH Reduced glutathione
Hb Hemoglobin
HETE Hydroxyeicosatetraenoic acid
12-HHT 12-Hydroxyheptadecatrienoic acid
HLE Human Leucozyte Elastase
IA2-A Tyrosinephosphatase antibody
IFN-c Interferon-c
IgG Immunoglobulin G
IgM Immunoglobulin M
IL Interleukin
IMG Immune globulin
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 293
Boswellic acids (BAs) belong to the active pharmacological principles of the oleo
gum resin from the trees of different Boswellia species. These trees are plants
typically found in the deserts, where the aborigines since thousands of years,
294 H.P.T. Ammon
scratch the bark and collect fluid, during dry periods. Burning the resin, the fumes
are used for disinfection, improvement of the scent of the air, as well as for cere-
monial and medical purposes. The resins are known as salai guggal (India),
frankincense (pure incense), incense and olibanum.
This chapter summarizes the present knowledge about medical history, phar-
macological active ingredients and therapeutical uses of the resin with special
impact on the anti-inflammatory mechanisms of boswellic acids.
The oldest written document, which mentions frankincense as a drug is the Papyrus
Ebers. In 1873, Moritz Fritz Ebers, professor of Egyptology, received a more than
20 m long papyrus from an Arab businessman [40] describing the medical use of
frankinsence in Egypt. In India, the therapeutic applications of the oleogum resin of
Boswellia serrata, called Salai guggal (SG), are already described in early
Ayurvedic textbooks (Charaka Samhita, 1st–2nd century AD and in Astangahrdaya
Samhita, seventh century AD).
Remedies containing preparations from frankincense (here Boswellia carterii)
were also prescribed by the famous physicians Hippocrates, Celsus, Galenus,
Dioskurides and others for the treatment of tumours, carcinomas, edemas, inflam-
matory diseases including diarrhoea, and diseases of the respiratory tract [40].
Olibanum, the resin of various Boswellia species (Table 13.1), was also known
as a remedy in Europe from ancient times till the beginning of the twentieth
century. Then it disappeared from the list of doctoral prescriptions since scientific
evidence for therapeutical efficacy was missing. Only when Singh and Atal [69] and
Ammon et al. [3] showed that an extract from resin of Boswellia serrata inhibited
inflammation in an animal model resp. formations of leukotrienes in an in vitro
model, the scientific community became interested and the first human pilot studies
were initiated. By now, preparations of the resin are widely used to treat a variety of
chronic inflammatory disorders.
The resin of Boswelli species consists of mucus, volatile oil and resin acids
(Table 13.2). However, the quantitative composition of these constituents varies
from species to species.
The resin acids contain pentacyclic and tetracyclic triterpenes. Among the
pentacyclic triterpenes, only some boswellic acids are responsible for many of the
pharmacological effects; but also tirucallic acids (TAs), from the tetracyclic triter-
penic acids, have been shown to be biologically active .
Pentacyclic Triterpenes: Büchele et al. [9] identified 12 different pentacyclic
triterpenes in the samples of Boswellia extracts (BEs) (Table 13.3). From these 12,
the chemical structures of the two most active boswellic acids (BAs) are shown in
Fig. 13.2.
The authors reported significant quantitative differences of pentacyclic triterpe-
nes between various species: A striking difference was observed in the content of
the boswellic acids, i.e. KBA and AKBA (Table 13.3). Recently Beisner et al. [5]
identified a new pentacyclic triterpene from Boswellia serrata, it was just one,
3a-acetyl-20(29)-lupene-24-oic acid and Verhoff et al. [80] observed biological
activity of a novel C(28)-hydroxylated lupeolic acid. Though BAs received most
attention through the group of pentacyclic triterpenes, some other, including lupeol,
exhibit pharmacological activity as well, which are also considered boswellic acids
[49].
Tetracyclic Triterpenes: Among the tetracyclic triterpenes, three TAs have been
identified: 3-oxotirucallic acid, 3-hydroxytirucallic acid and 3-acetoxytirucallic
acid. Other resin compounds with pharmacological activities are: betulinic acid,
epi-lupeol, lupenoic acid, 1-ursene-2-diketone-incensole acetate, isoincensole and
isoincensole acetate and as well as terpenes that can be found in the volatile oil.
The first scientific publication with the title “Analgesic effect of the oleogum resin
of Boswellia serrata Roxb”. by Karr and Mennon appeared 1969 [31]. Singh and
Fig. 13.1 Boswellia serrata Roxb. Desert of Radschastan, India, taken by the author
Fig. 13.4 Inhibition of 6-keto-PGF1a formation in human platelets by an alcoholic extract of salai
guggal (SG). 100 % = 277 pg 6-keto-PGF1a/107 cells. Number of experiments are given in
parentheses [3]
Until 1991, there was no drug known which could predominantly inhibit the
synthesis of leukotrienes despite the need for such compounds to treat diseases
where leukotrienes have a major impact. It was the publication of Ammon et al. [3]
that showed that BEs not only affected formation of COX products, but to a much
larger extend also inhibited LTB4 synthesis. This publication and a later paper [50],
indicating that certain BAs are responsible for this effect, received large attention by
the scientific community.
Boswellic Extracts: After the stimulation of leukotriene synthesis with the calcium
ionophore A 231876 in polymorphmononuclear neutrophils (PMN)—which con-
tained 5-LO but no COX the same extract that was used for the studies on pros-
taglandin synthesis [3] in a concentration-dependent manner inhibited synthesis of
leukotriene B4 (LTB4) and 5-hydroxyeicosatetraenoic acid (5-HETE) (a metabolite
of the 5-LO cascade) formation at concentrations between 10 and 80 lg/ml
(Fig. 13.5). In this case, 50 % inhibition occurred at a concentration of 30 lg/ml,
indicating that the inhibitory action of the extract was significantly more pro-
nounced than its effect on COX-1 [3].
Boswellic Acids: Concerning the actions of BAs, Safayhi et al. reported in 1992
BAs to be specific, non-redox inhibitors of 5-LO. In this study, isomers of (a- and
b-) of BAs and their acetyl derivatives were isolated from the oleogum resin of BS.
It was observed that BAs partly decreased the formation of LTB4 in
calcium-ionophore-stimulated PMN in a concentration-dependent manner. AKBA
was most effective with an IC50 value of 1.5 µM.
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 301
Oxygen radicals which are formed in PMN through the action of leukotrienes are
also involved in cartilage destruction in rheumatoid arthritis. Heil et al. [27] studied
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 303
White blood cells play an important role in the defence system of the body.
Granulocytes, monocytes, macrophages and lymphocytes cover the non-specific as
well as the humoral and cellular defence. However, under certain conditions, they
are also closely related to inflammatory autoimmune disorders. Humoral and cel-
lular defence have also been the subject of studies with BEs and BAs.
Humoral defence is one of the important measures of the body against infectious
diseases. It is related to the activation of B lymphocytes. After contact with anti-
gens, B cells differentiate to plasma cells, which then produce antibodies belonging
to the family of immunoglobulins.
Antibody Titres
Boswellic Acids: Humoral antibody synthesis in mice treated with sheep ery-
throcytes was studied by Sharma et al. [62] by determining the hemagglutinating
antibody titers in the serum. Here, primary antibody production (after a first antigen
injection) and secondary antibody production (after a second injection) were tested.
It was found that a single dose of a mixture of BAs (50–200 mg/kg)—as isolated by
Singh et al. [70] on the day of sensitization produced a dose related reduction
(10.4–32.8 %) in primary hemagglutinating antibody titers on day 4. Significant
reduction in antibody production was obtained with 100 and 200 mg/kg doses. On
the other hand the secondary antibody titers were significantly enhanced at lower
doses, the effect being more prominent at 50 mg/kg. Azathioprine was administered
as a reference compound (200 mg/kg p.o.) following the same schedule and
resulted in only 10.4 % inhibition of primary antibody synthesis and had no effect
on the secondary antibody production.
Usually, the second injection of antigens causes earlier antibody production with
higher affinity and is mainly due to immunoglobulin G (IgG).
Using the technique of complement fixing, a method for analysing antigen and
antibody titers, oral administration of a mixture containing BAs for 5 days around the
time of immunization resulted in a significant decrease in primary and secondary
complement fixing antibody titers at 100 mg/kg [62]. On the contrary, when a BA
mixture (25–100 mg/kg) was given orally for 5 days around immunization a marked
increase (15.38–26.92 %) in antibody production on day +7 was observed. The effect
was more pronounced at a dose of 25 mg/kg than at 50 or 100 mg/kg. The secondary
antibody titers were only marginally increased. Azathioprine treatment (100 mg/kg)
had no significant effects on primary as well as on secondary antibody titers.
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 305
In mice, in which treatment with BAs was initiated 7 days prior to immuniza-
tion, BAs (25–100 mg/kg) elicited a dose related increase (37.93–63.79 %) in the
primary humoral response without significantly affecting the expression of the
secondary response. Levamisole (2.5 mg/kg, p.o.), an immunopotentiating agent,
displayed only a 25 % increase in primary and a 6.66 % increase in secondary
antibody titers.
Immunoglobulins
Boswellic Extract As far as low doses of BEs are concerned, the data received
with antibody titers finds their equivalent when immunoglobulins were determined
in the blood of sensitized mice. Previously, it was shown by Khajuria et al. [33] that
oral administration of a biopolymeric fraction (BOS 2000) from B. serrata (1–
10 mg/kg) elicited a dose related increase in the delayed hypersensitivity reaction
(early 24 h and delayed 48 h) in mice. It also stimulated the immunoglobulin M
(IgM) and IgG titers expressed in the form of plaques (PFC) and the complement
fixing antibody titre.
Gupta et al. [26] studied a possible immunological adjuvant effect of BOS 2000 on
specific antibody and the cellular response to ovalbumin in mice. Mice were
immunized s. c. with ovalbumin 100 µg and received BOS 2000, 80 µg on days 1
and 15. Two weeks later, BOS 2000, 80 µg corresponding to about 3.5 mg/kg bw
significantly increased IgG, IgG1 and IgG2a antibody levels in the serum compared
to the ovalbumin group.
At a dose of 80 µg BOS 2000, there was also a significant increase of lym-
phocytes CD4/CD8 and CD80/CD 86 analyzed in spleen cells and with cytokines
(IL-2 and IFN-c) profiles in the spleen cell culture supernatant. From their data, the
authors conclude, that BOS 2000 seems to be a promising balanced Th1- and Th2
lymphocytes directing immunological adjuvant which can enhance the immuno-
genicity of vaccines.
Thus, regarding the humoral defence in the in vivo experiments, it appears that
BAs have a dual effect on antibody titers: at lower doses, there is an increased
formation, whereas at higher doses, BAs may even have inhibitory effects. Whether
or not this data is of relevance for humans remains to be established. And the
question remains: what is a high and what is a low-dose regarding humans.
Their functions are organized through the crosstalking among related cells via
cytokines produced and released from various leucocytes. Numerous authors have
dealt with the question whether or not extracts from the Boswellia resin, its volatile
oil, and/or boswellic acids would affect the activity of white blood cells and whether
or not such effects could be attributed to an interaction with the system of cross
talking between white blood cells by cytokines.
Boswellic Extracts and Essential Oils: In their studies on lymphocyte transfor-
mation, Badria et al. [4] used an assay with lymphocytes isolated from venous
human blood. In this study, a methylene chloride extract from the oleogum resin of
Boswellia carterii in the presence of phytohemaglutinin (PHA) or concanavalin A
(CON A) at 1 mg/ml stimulated lymphocyte transformation by 90 %
(EC50 = 0.55 mg/ml). Several compounds from the essential oil were also bio-
logically active. The different BAs and TAs tested, including acetyl-b-BA,
acetyl-a-BA, 3-oxo-TA, AKBA, b-BA, 3-hydroxy-TA and KBA, showed a similar
activity with EC50 values from 0.001 to 0.005 lM.
Mikhaeil et al. [41], who studied the chemical composition of frankincense oil,
also reported that the oil exhibited strong immunostimulant activity (90 %) of
lymphocyte transformation, when assessed in a lymphocyte proliferation assay.
From these studies, it may be concluded, that a variety of components of an extract
from Boswellia resins are effective in stimulating lymphocyte transformation and
thus can contribute to cellular defence.
Boswellic Acids: Sharma et al. [62] reported that, if spleen cells from
non-immunized mice were used, a mixture of various BAs in the range of 1.95–
125.0 lg/ml showed no spontaneous mitogenic activity and the cell viability was
comparable to controls. However, when, the test, was performed in the presence of
mitogen stimulating lipopolysaccharides (LPS, PHA, CON A and alloantigen), a
concentration-dependent inhibition of lymphocyte proliferation by BAs was
observed.
The effect of a mixture of BAs on phagocytosis was also studied by Sharma et al.
[62]. Preincubation of peritoneal macrophages with different concentrations of BAs
(1.95–125 lg/ml) resulted in an enhanced phagocytotic function of adherent
macrophages with a maximal effect occurring at 62.25 lg/ml.
The studies discussed so far have been performed in vitro. They showed stim-
ulating and inhibitory effects of BAs and BEs, respectively, employing different
concentrations. At present, the data is difficult to interpret and transferred to the
human situation.
In an in vivo experiment studying the anti-arthritic activity of boswellic acids
employing bovine serum albumin (BSA) induced arthritis, Sharma et al. [61]
observed that oral administration of a mixture of BAs (25, 50 and 100 mg/kg/day)
reduced the population of leucocytes when BSA was injected into the knee, and
changed the electrophoretic pattern of the synovial fluid proteins. The local injec-
tion of BAs (5, 10 and 20 mg) into the knee 15 min prior to BSA challenge also
reduced infiltration of leucocytes into the knee joint and inhibited the migration
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 307
properties of PMN in vitro. As discussed above some BAs are inhibitors of 5-Lo.
So, the mechanism of action of BAs on leucocyte infiltration could be the inhibition
of LTB4—synthesis, i.e. reducing their chemotactic action.
NFjB plays a key role for the function of cytokines in their crosstalk between
immune competent cells. NFjB, which is usually present in the cytosol, is produced
by neutrophil granulocytes. This compound regulates the release of cytokines from
various white blood cells being responsible for proliferation, activation and function
of these cells.
As far as possible effects of BEs are concerned, AKBA, as one of their pharma-
cological active compounds, has turned out to be a natural inhibitor of NFjB [16].
Thus, AKBA applied in vivo in mice (100 lmol/kg) for 1 week inhibited the NFjB
activation. Suppression of NFjB and NFjB-regulated gene expression by AKBA is
also reported by Takada et al. [77].
When AKBA was given systemically or locally in a mouse model with psoriasis,
the signalling action of NFjB and subsequent NFjB-dependent cytokine produc-
tion by macrophages was significantly suppressed. This was associated with pro-
found improvement of the psoriasis disease activity score [82]. As far as the
mechanism of action on NFjB is concerned, Syrovets et al. [76] reported that
acetyl-boswellic acids inhibit constitutively activated NFjB signalling by inter-
cepting the IjB-kinase activity in an in vitro model of PC-3 prostate cancer cells.
However, inhibition of NFjB is not only due to an action of BAs in extracts from
Boswellia species. Previously, Moussaieff et al. [45] reported that incensole acetate,
a compound isolated from Boswellia resins, also inhibits NFjB activation. This
action was combined with an anti-inflammatory effect in the inflamed mouse paw
model.
Cytokines
Interleukines/IFN-c
Interleukins belong to the group of cytokines. There are pro- and anti-inflammatory
interleukins produced by macrophages and T cells following the recognition of a
pathogen.
Chervier et al. [13] studied the effect of an extract from Boswellic carterii on the
production of TH-1 and TH-2 cytokines by murine splenocytes. The use of an
extract with sesame oil as solvent resulted in a dose-dependent inhibition of IL-2
and IFN-c and a dose-dependent potentiation of IL-4 and IL-10. Gayathri et al. [19]
observed that a crude methanolic extract from BS and 12-ursine-2-diketone, a pure
compounds of BS, inhibited TNF-a, IL-1 B and IL-6 in cultured Peripheral Blood
Mononuclear Cells (PBMC). Observations on TH1/TH2 cytokines also revealed
marked down regulation of IFN-c and IL-12, whereas IL-4 and IL-10—which are
anti-inflammatory interleukins—was upregulated upon treatment with a crude
extract and the pure compound 1-ursene-2-diketone. This indicates that both are
capable of carrying out anti-inflammatory activities at sites where chronic inflam-
mation is present by switching off the proinflammatory cytokines.
Employing an acetone extract from Boswellia carterii Birdw. Fan et al. [18], in
an adjuvant arthritis model in lewis rats 900 mg/kg for 10 consecutive days,
observed significant decrease of arthritic scores. This was associated with sup-
pression of local tissue IL-1B and TNF-a. On the other hand, in a study of Khajuria
et al. [33], the authors demonstrated that oral administration of 1–10 mg/kg of a
polymeric fraction from BS (BOS 2000) increased levels of IL-4, IFN-c and TNF-a
in the serum.
The data from this chapter, covering cytokines, suggest that low doses of
Boswellia preparations increase production of proinflammatory cytokines,
whereas high doses are even suppressive. In other words, low doses of BEs
may stimulate cellular defence f.i. in case of infections and high doses may be
useful in defeating the proinflammatory actions of cytokines in autoimmune
inflammatory disorders.
Fig. 13.6 Effect of boswellic extract (150 mg/kg i.p. for 10 days) on blood glucose levels of
MLD-STZ-treated mice (mean ± SE; n = 4) [64]
310 H.P.T. Ammon
Fig. 13.7 Effect of Boswellic extract (150 mg/kg i.p. for 10 days) on proinflammatory cytokines
in the serum of MLD-STZ-treated mice. C control (mean ± SE; n = 4) [64]
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 311
Resins in general protect the stems of trees from any microbial attack.
Raja et al. [48] studied the antimicrobial activities of various Boswellic acids
against 112 pathogenic bacterial isolates including ATCC strains. Here, AKBA
exhibited the most potent antibacterial activity showing a Minimal Inhibitory
Concentration (MIC) range of 2–8 lg/ml against the entire Gram-positive bacterial
pathogens tested. It exhibited a concentration-dependent toxicity of Staphylococcus
aureus ATCC 29213 up to 8 MIC. The antibacterial mode of action of AKBA
probably occurred via disruption of the microbial membrane structure.
In another study, methanolic and aqueous extracts from Boswellia dalziellia also
showed a broad spectrum of inhibitory activity against bacteria, both Gram positive,
Gram negative and fungi [2].
Serratol, a diterpene isolated from the gum resin of Boswellia serrata, was
previously tested by Schmidt et al. [57] for its antiprotozoal activity. It was found to
be active against Tripanosoma brucie, Rhode siense (sleeping sickness) and
Plasmodium falciparum (Tropical Malaria).
Essential oils of the gum resin from different Boswellia species including
Boswellia carterii (Somalia), Boswellia papyrifera (Ethiopia), Boswellia serrata
(India) and Boswellia rivae (Ethiopia) were tested by Camardes et al. [11] and
showed antimicrobial activity.
So far, only extracts from the gum resin of Boswellia serrata have been used for
clinical trials. Studies with isolated compounds are still missing. The studies focus
on chronic inflammatory diseases.
In modern pharmacology, the therapeutic effects of drugs are closely related to their
pharmacokinetic properties. However, as far as extracts from herbal medicine are
concerned, it is impossible to establish pharmacokinetic data since they contain a
312 H.P.T. Ammon
Table 13.5 Antimicrobial and antiparasitical effects of extracts, essential oils, and chemical
constituents from different Boswellia species
Extracts
Methanolic extract (stem Broad spectrum of gram positive and gram negative [2]
bark) bacteria and fungi
B. dalzielli
Methanolic extract Gram positive multiresistent Staphylococcus strains [42]
B. ameero
B. elongata
Boswellic extract Influenza virus A [43]
B. ameero
B. elongata
Methanolic extract Hepatitis virus C [29]
B. carterii
Methanolic extract from Musceocideal [23]
stem bark
Essential oils
Essential oil different B. Gram positive bacteria, gram negative bacteria, antifungal [11]
species
Essential oil B. riva Candida albicans [56]
Chemical constituents
Serratol (diterpene from B. Tripanosoma brucei rhodesiense (sleeping sicknes) [57]
serrata) Plasmodium falsiparum (tropical Malaria)
AKBA Staphylococcus aureus, gram positive bacterial pathogens [48]
variety of compounds which are involved in the final therapeutic actions. Moreover,
their contents vary from species to species. Thus, the only possibility for a rough
standardization is to determine one or more components of an extract where sig-
nificant pharmacological effects can be expected. At present, in case of BEs, these
are KBA and AKBA. Studies with isolated KBA and AKBA in humans do not exist
yet. (Table 13.5)
In an open uncontrolled trial with 12 healthy male volunteers [63], a single oral
dose of a commercial product of an extract out of gum resin from Boswellia serrata
(WokVel™) containing 333 mg was given after standard breakfast. The extract
contained 6.44 % KBA, 2 % AKBA, 18.51 % b-BA, 8.58 % 3-O-acetyl-b-BA,
6.93 % a-BA and 1.85 % 3-O-acetyl-aBA. In this study, only the concentration of
KBA in the plasma was measured (Fig. 13.8). As shown in Table 13.6 maximum
concentration of KBA was found after 4.5 h 2.72 µM. This concentration is in the
range of the IC50 of KBA for the inhibition of leukotriene synthesis in vitro. In this
study, the elimination half life (t1/2b) was 5.97 h. This suggests that when the
above treatment is given every 6 h, the steady state plasma concentration of KBA is
reached after approximately 30 h.
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 313
Fig. 13.8 Mean plasma concentration of 11-keto-b-boswellic acid versus time after oral
administration of 333 mg Boswellia serrata extract WokVel™ containing 6.44 % KBA [63]
Boswellia. Twelve healthy subjects fasted 10 h before and 4 h after drug admin-
istration (group A). A second group (B) received a high-fat meal together with the
drug. The volunteers swallowed a single dose of 3 capsules with 282 mg (total of
786 mg) extract, containing 143.4 mg b-BA, 103.71 mg a-BA, 82.71 mg
acetyl-b-BA, 48.12 mg KBA, 28.71 mg AKBA and 26.25 mg acetyl-a-BA. The
time course of the plasma concentrations of the most active boswellic acids, i.e.
KBA and AKBA, was dramatically different between fasted and high-fat meal
volunteers. The calculated pharmacokinetic parameters are shown in Table 13.7.
It is obvious that in the high-fat meal group Cmax of KBA is about 2.7 times and
AKBA 4.8 times higher than in the fasting group. This can be explained by the
lipophilic character of KBA and AKBA. Consequently, BEs should be taken
together with food.
13.6.1.2 Absorption
Extract preparations from the resin of Boswellia species, in most cases, are
administered by oral route. Compared to the concentrations of BAs, in extract
preparations their concentrations in the blood after oral application are very low.
This holds true, especially for the most active BA, i.e. AKBA. Krüger et al. [37]
explain this phenomenon to be due to poor permeability of AKBA and also
moderate absorption of KBA.
13.6.1.3 Distribution
13.6.1.4 Metabolism
In rat liver microsomes, human liver microsomes and hepatocytes, Krüger et al.
[36] found that KBA but not AKBA undergoes extensive phase I metabolism.
Oxidation to hydroxylated metabolites is the principal metabolic route. In vitro,
KBA yielded metabolic profiles similar to those obtained in vivo in rat plasma and
liver, whereas no metabolites of AKBA could be identified in vivo. Unexpectedly,
AKBA is not deacetylated to KBA.
Extracts from the gum resin of BS are frequently used topically in ointment
preparations for skin diseases. Singh et al. [71] reported anti-inflammatory activity
of BAs through this route in different acute and chronic models of inflammation
such as arachidonic acid- and croton-oil-induced mouse ear edema,
carrageenan-induced rats paw edema and adjuvant-induced arthritis in rats. The
results of the study revealed that the antiphlogistic effect observed through this
route is in accordance with the study conducted with the systemic application.
From the preclinical studies it is reasonable to conclude that BEs and their active
constituents will be effective in inflammatory disorders, since they inhibit the
responsible related factors, i.e. products of the arachidonic acid cascade, especially
leukotrienes, members of the complement system—and especially factors of the
316 H.P.T. Ammon
As early as 1986, Singh and Atal [69] reported that in an arthritic model in rats a
mixture of boswellic acids showed anti-inflammatory activity and Sharma et al. [61]
observed that in another arthritis model a mixture of BAs reduced infiltration of
leucocytes into an arthritic knee. Very recently, Umar [79] studied the effect of an
extract from Boswellia serrata in the collagen-induced arthritis model in rats on
most of the inflammatory parameters discussed. BE was administered at doses of
100 and 200 mg/kg body weight once daily for 21 days. The effects of the treat-
ment in rats were assessed by biochemical parameters (articular elastase, LPO,
GSH, catalase, SOD and NO), inflammatory mediators (IL-1B, IL-6, TNF-a, IL-10,
IFN-c and PGE2), and histopathology in joints. In this study, BE was effective
bringing significant changes to all the tested parameters (articular elastase, LPO,
GSH, catalase, SOD and NO). Oral administration of BE resulted in significantly
reduced levels of inflammatory mediators (IL-1B, IL-6, TNF-a, IFN-c and PGE2),
and increased levels of IL-10. The protective effects of BE against RA were also
evident from the decrease in arthritis scores and bone histology. On the basis of
these studies and traditional experiences, a variety of clinical studies have been
initiated including subjects suffering from rheumatoid arthritis and osteoarthritis.
Rheumatoid Arthritis
product H15™ (produced by Gufic Ltd. Mumbai, India), when compared to the
placebo group (p < 0.05). Unfortunately, not all of these studies could be
re-examined by the author of this chapter as they were not published. As a con-
sequence, the quality and the outcome of these studies were critiqued by the
German Society of Rheumatology in 1998. The arguments are mainly based on a
study of Sander et al. [55]. In this multicentre controlled trial, the authors studied
the effect of H 15™ versus placebo over a period of 12 weeks in 37 outpatients
with rheumatoid arthritis and chronic polyarthritis being under constant therapy
with steroids and disease-modifying antirheumatic drugs. The patients received 9
tablets of H 15™ (3600 mg) or placebo daily in addition to their previous therapy.
Doses of NSAIDs could be adjusted on demand. Efficacy parameters were the index
for swelling and pain, ESR and CRP. Pain and NSAID doses were documented at
the beginning and at 6 and 12 weeks after initiation. In this study, treatment with
H15™ resulted in no measurable effect. However, this study suffers from the
drawback that the effect of the BE alone in comparison to standard therapy was not
tested. It can be assumed that administration of H15™ to patients who are being
already treated with steroids and patients with basic therapy will not experience an
additional effect.
Osteoarthritis
Ulcerative Colitis
Chronic Colitis
This disease was characterized by the authors [25] as vague lower abdominal pain,
bleeding per rectum with diarrhoea and palpable tender descending and sigmoid
colon. Its pathophysiology seems to be different from that of ulcerative colitis.
Boswellic Extract: In this study, 30 patients, 17 males and 13 females age 18–
48 years, were included. Twenty patients were given a preparation of the oleogum
resin of Boswellia serrata (S-Compound™) containing KBA 0.63 %, AKBA
0.7 %, acetyl-b-BA and b-BA 1.5 % (900 mg daily divided in three doses for
6 weeks) and ten patients receiving sulfasalazine (3 mg daily divided in three doses
for 6 weeks) served as controls. Out of the 20 patients treated with Boswellia
oleogum resin, 18 patients showed an improvement in one or more of the following
parameters: stool properties, histopathology as well as scanning electron micro-
scopy, Hb, serum iron, calcium, phosphorus, proteins, total leukocytes, and
eosinophils.
Collagenous Colitis
Crohn’s Disease
Since ancient times (see Chap. 2) respiratory symptoms are treated with prepara-
tions of olibanum/frankincense. This still holds for bronchitis including cough and
expectoration. A special case is bronchial asthma.
Bronchial asthma is a chronic inflammatory condition characterized by bron-
chial hyper-responsiveness and reversible airways obstruction.
Increased production of leukotrienes both during episodes of asthma and in
patients with stable asthma was shown by Chanarin and Johnston [14]. Bronchial
asthma is also a case of autoimmune diseases.
Boswellia Extracts: In a double-blind, placebo-controlled trial with asthma
patients Gupta et al. [24] tested forty patients with a mean duration of bronchial
asthma of 9.58 ± 6.07 years. They received a preparation of oleogum resin of
Boswellia serrata (S-Compound™) of 300 mg three times daily over a period of
6 weeks. In this study, 70 % of the patients showed improvement of the disease
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 321
Taking into account the wide use of oleogum resin from different Boswellia species
in ancient times and nowadays, side effects do not appear to be a critical point. This
is also the outcome of the published material. Most of the side effects, if at all, are
related to the gastrointestinal tract. In detail:
Gastrointestinal Tract: In the study using S-Compound™ [22], dyspeptic
symptoms in form of pain in abdomen, distension of abdomen, sour eructation and
loss of appetite were reported in 8 % while 60 % of patients treated with Ibuprofen
had dyspeptic symptoms.
In a further trial with S-Compound™, two from 40 patients complained about
epigastric pain, hyperacidity, and nausea [24]. In a study dealing with ulcerative
colitis [23], 6 out of 34 patients reported retrosternal burning, nausea, fullness of
abdomen, epigastric pain and anorexia.
Böker and Winking [7], studying the effect of H15™, reported that some
patients developed nausea and vomiting. The side effects were reversible after
omission of the treatment. Comparing the effect of H15™ (44 patients) with
mesalazine (39 patients) suffering from Crohn’s disease), Gerhardt et al. [20]
reported no drug related side effects in the H15™ group but 4 patients treated with
mesalazine suffered from headache and gastrointestinal symptoms. In the study of
Gupta et al. [25], out of 20 patients treated with Boswellia gum resin, only 2
patients complained of heartburn and Streffer et al. [75] reported that the prepa-
ration H15™ was well tolerated, only some gastrointestinal complains were
observed. BE (15 patients) was also well tolerated in the study of Kimmatkar et al.
[34] exept for minor gastrointestinal reactions. In the study of Sontakke et al. [72],
one patient out of 33 receiving BE complained about diarrhoea and abdominal
cramps.
Boswellic extracts in higher doses are used in treatment of cerebral edemas
caused by brain tumours. In a prospective, randomized, placebo-controlled
double-blind pilot study [35], where 22 patients with brain tumours received
4200 mg of a BE (H15™) or placebo, diarrhoea grade 1–2 occurred in 6 patients of
the BE group. Other side effects were the same as in the placebo group.
Respiratory Tract: There is a case report from a patient who complained about
asthma symptoms after exposure to fume of incense in the church [46].
Skin: In an other case report Acebo et al. [1] describe a patient suffering from
allergic contact dermatitis using an extract from Boswellia serrata in a naturopathic
cream.
In two patients being treated with H15™ for brain edema, Böker and Winking
[7] described skin irritations. The side effects were reversible after omission of the
treatment.
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 323
References
1. Acebo E, Raton JA, Saulua S, Eizaguirre X, Trébol J, Perez JLD (2004) Allergic contact
dermatitis from Boswellia serrata extract in a naturopathic cream. Contact Dermat 51:91
2. Adelakun EA, Finbar EA, Agina SE, Makinde AA (2001) Antimicrobial activity of Boswellia
dalzielii stern bark. Fitoterapia 72:822–824
3. Ammon HP, Mack T, Singh GB, Safayhi H (1991) Inhibition of leukotriene B4 formation in
rat peritoneal neutrophils by an ethanolic extract of the gum resin exudates of Boswellia
serrata. Planta Med 57:203–207
4. Badria FA, Mikhaeil BR, Maatooq GT, Amer MM (2003) Immunmodulatory terpenoids from
the oleogum resin of Boswellia carterii Bordwood. Z Naturforsch 58:506–516
5. Beisner K, Büchele B, Werz U, Simmet T (2003) Structural analysis of 3-a-acetyl-20(29)-
lupene-24-oic acid, a novel pentacyclic triterpene isolated from the gum resin of Boswellia
serrata, by NMR spectroscopy. Magn Reson Chem 41:629–632
6. Blain EJ, Ali AY, Duance VC (2010) Boswellia frereana (frankincense) suppresses
cytokine-induced matrix metalloproteinase expression and production of proinflammatory
molecules in articular cartilage. Phytother Res 24:905–912
7. Böker DK, Winking M (1997) Die Rolle von Boswellia-Säuren in der Therapie maligner
Gliome. Dtsch Ärztebl 94:B-958–B-960
8. Büchele B, Simmet T (2003) Analysis of 12 different pentacyclic triterpenic acids from
frankincense in human plasma by high performance liquid chromatography and photodiode
array detection. J Chromatogr B 795:355–362
9. Büchele B, Zugmaier W, Simmet T (2003) Analysis of pentacyclic triterpenic acids from
frankincense gum resins and related phytopharmaceuticals by high-performance liquid
chromatography. Identification of lupeolic acid, a novel pentacyclic triterpene. J Chromatogr B
791:21–30
10. Buvari PG (2001) Wirksamkeit und Unbedenklichkeit der H15 Ayurmedica-Therapie bei
chronisch entzündlichen Erkrankungen. Dissertation, University of Mannheim-Heidelberg
324 H.P.T. Ammon
32. Kela SL, Ogunsusi RA, Ogbogu VC, Nwude N (1989) Screening of some Nigeria plants for
molluscicidal activity. Rev Elev Med Vet Pays Trop 42:195–202
33. Khajuria A, Gupta A, Suden P, Singh S, Malik F, Singh J et al (2008) Immunmodulatory
activity of biopolymeric fraction BOS 2000 from Boswellia serrata. Phytother Res 22:340–
348
34. Kimmatkar N, Thawani V, Hingorani L, Khiyani R (2003) Efficacy and tolerability of
Boswellia serrata extract in treatment of osteoarthritis of knee—a randomized double blind
placebo controlled trial. Phytomedicine 10:3–7
35. Kirsten S, Treier M, Wehrle SJ, Becker G, Abdel-Tawab M, Gerbeth K et al (2011) Boswellia
serrata acts on cerebral edema in patients irradiated for brain tumors. Cancer 117:3788–3795
36. Krüger P, Daneshfar R, Eckert GP, Klein J, Volmer DA et al (2008) Metabolism of boswellic
acids in vitro and in vivo. Drug Metabol Distpos 36:1135–1142
37. Krüger P, Kanzer J, Hummel J, Fricker G, Schubert-Zsilavecz M, Abdel-Tawab M (2009)
Permeation of Boswellia extract in the Caco-2 model and possible interactions of its
constituents KBA and AKBA with OATP1B3 and MRP2. Eur J Pharm Sci 36:275–284
38. Madisch A, Miehlke S, Eichele O, Bethke B, Mrwa J, Kuhlisch E et al (2005) Boswellia
serrata Extrakt bei kollagener Kolitis – eine randomisierte, placebo-kontrollierte, doppelblinde
Multicenterstudie. Dtsch Ges Verdau Stoffw Erkr. Gastroenterol 43(Suppl. Z):P061
39. Martelli L, Berardesca E, Martelli M (2000) Topical formulation of a new plant extract
complex with refirming properties. Clinical and non-invasive evaluation in a double-blind
trial. Int J Cosmet Sci 22:201–206
40. Martinez D, Lohs K, Janzen J (1989) Weihrauch und Myrrhe. Kulturgeschichte und
wirtschaftliche Bedeutung. Botanik, Chemie, Medizin. Wissenschaftliche Verlagsgesellschaft,
Stuttgart
41. Mikhaeil BR, Maatooq GT, Badria FA, Amer MM (2003) Chemistry and immunomodulatory
activity of frankincense oil. Z Naturforsch C 58:230–238
42. Mothana RA, Lindequist U (2005) Antimicrobial activity of some medicinal plants of the
island Sogotra. J Ethnopharmacol 96:177–181
43. Mothana RA, Mentel R, Reiss C, Lindequist U (2006) Phytochemical screening and antiviral
activity of some medicinal plants from the island Sogotra. Phytother Res 20:298–302
44. Mothana RA (2011) Anti-inflammatory, antinociceptive and antioxidant activities of the
endemic Sogotraen Boswellia elongata Balf. F. and Jatropha unicostata Balf. F. in different
experimental models. Food Chem Toxicol 49:2594–2599
45. Moussaieff A, Shohami E, Kashman Y, Fride E, Schmitz ML, Renner F et al (2007)
Incensolate acetate, a novel anti-inflammatory compound isolated from Boswellia resin,
inhibits nuclear factor-kappa B activation. Mol Pharmacol 72:1657–1664
46. O’Connor TM, Cusack R, Lauders S, Bredin CP (2014) Holy Saturday asthma. BMJ Case
Rep doi:10.1136/bcr-2014-203861
47. Poeckel D, Tausch L, Kather N, Jauch J, Werz O (2006) Boswellic acids stimulate arachidonic
acid release and 12-lipoxygenase activity in human platelets independent of Ca2+ and
differentially interact with platelet-type 12-lipoxygenase. Mol Pharmacol 70:1071–1078
48. Raja AF, Ali F, Khan IA, Shawl AS, Arora DS (2011) Acetyl-11-keto-b-boswellic acid
(AKBA); targeting oral cavity pathogens. BMC Res Notes 4:406
49. Reddy KP, Singh AB, Puri A, Srivastava K, Narender T (2009) Synthesis of novel triterpenoid
(lupeol) derivatives and their in vivo antihyperglycemic and antidyslipidemic activity. Bioorg
Med Chem Lett 19:4463–4466
50. Safayhi H, Mack T, Sabieraj J, Anazodo MI, Subramanian LR, Ammon HP (1992) Boswellic
acids: novel, specific, nonredox inhibitors of 5-lipoxygenase. J Pharmacol Exp Ther
261:1143–1146
51. Safayhi H, Sailer ER, Ammon HP (1995) Mechanism of 5-lipoxygenase inhibition by
acetyl-11-keto-beta-boswellic acid. Mol Pharmacol 47:1212–1216
52. Safayhi H, Rall B, Sailer ER, Ammon HP (1997) Inhibition by Boswellic acids of human
leukocyte elastase. J Pharmacol Exp Ther 281:460–463
326 H.P.T. Ammon
53. Sailer ER, Subramanian LR, Rall B, Hoernlein RF, Ammon HP, Safayhi H (1996)
Acetyl-11-keto-beta-boswellic acid (AKBA): structure requirements for binding and
5-lipoxygenase inhibitory activity. Br J Pharmacol 117:615–618
54. Sailer ER, Schweizer S, Boden SE, Ammon HP, Safayhi H (1998) Characterization of an
acetyl-11-keto-beta-boswellic acid and arachidonate-binding regulatory site of 5-lipoxygenase
using photoaffinity labeling. Eur J Biochem 256:364–368
55. Sander O, Herborn G, Rau R (1998) Is H15 (extract of Boswellia serrata “incense”) an
efficient supplementation to established drug therapy of rheumatoid arthritis? Results of a
double-blind pilot trial. Z Rheumatol 57:11–16
56. Schillaci D, Arizza V, Dayton T, Camarda L, Di SV (2008) In vitro anti-biofilm activity of
Boswellia spp. Oleogum resin essential oils. Lett Appl Microbiol 47:433–438
57. Schmidt TJ, Kaiser M, Brun R (2011) Complete structural assignment of serratol, a
cembrane-type diterpene from Boswellia serrata and evaluation of its antiprotozoal activity.
Planta Med 77:849–850
58. Schrott E, Laufer S, Lämmerhofer M, Ammon HPT (2014) Extract from gum resin of
Boswellia serrata decreases IA2-antibody in a patient with “Late onset Autoimmune Diabetes
of the Adult” (LADA). Phytomedicine 21:786
59. Sengupta K, Alluri KV, Satish AR, Mishra S, Golakoti T, Sarma KV et al (2008) A double
blind, randomised, placebo controlled study of the efficacy and safety of 5-Loxin for treatment
of osteoarthritis of the knee. Arthritis Res Ther 10:R85
60. Sengupta K, Krishnaraju AV, Vishal AA, Mishra A, Trimurtulu G, Sarma KV et al (2010)
Comperative efficacy and tolerability of 5-Loxin and Aflapin. Against osteoarthritis of the
knee: a double blind, randomized, placebo controlled clinical study. Int J Med Sci 7:366–379
61 Sharma ML, Bani S, Singh CB (1989) Antiarthritic activity of boswellic acids in bovine serum
albumin (BSA)-induced arthritis. Int J Immunopharmacol 11:647-652
62. Sharma ML, Kaul A, Khajuria A, Singh S, Singh GB (1996) Immunomodulatory activity of
boswellic acids (pentacyclic triterpene acids) from Boswellia serrata. Phytother Res 10:
107–112
63. Sharma S, Thawani V, Hingorani L, Shrivastava M, Bhate VR, Khiyani R (2004)
Pharmacokinetic study of 11-keto-beta-boswellic acid. Phytomedicine 11:255–260
64. Shehata AM, Quintanilla-Fend L, Bettio S, Singh CB, Ammon HP (2011) Prevention of
multiple low-dose streptozotocin (MLD-STZ) diabetes in mice by an extract from gum resin of
Boswellia serrata (BE). Phytomedicine 18:1037–1044
65. Shehata AM, Quintanilla-Fend L, Bettio S, Jauch J, Scior T, Scherbaum WA, Ammon HPT
(2015) 11-Keto-b-Boswellic acids prevent development of autoimmune reactions, insulitis and
reduce hyperglycemia during induction of multiple low dose streptozotocin (MLD-STZ)
diabetes in mice. Horm Metab Res 47:463–469
66. Siemoneit U, Hofmann B, Kather N, Lamkemeyer T, Madlung J, Franke L et al (2008)
Identification and functional analysis of cyclooxygenase-1 as a molecular target of Boswellic
acids. Biochem Pharmacol 75:503–513
67. Siemoneit U, Pergola C, Jazzar B, Northoff H, Skarke C, Jauch J, Werz O (2009) On the
interference of Boswellic acids with 5-lipoxygenase: mechanistic studies in vitro and
pharmacological relevance. Eur J Pharmacol 606:246–254
68. Siemoneit U, Koeberle A, Rossi A, Dehm F, Verhoff M, Reckel S et al (2011) Inhibition of
microsomal prostaglandin E2 synthase-1 as a molecular basis for the anti-inflammatory actions
of Boswellic acids from frankincense. Br J Pharmacol 162:147–162
69. Singh GB, Atal CH (1986) Pharmacology of an extract of salai guggal ex Boswellia serrata, a
new non-steroidal anti-inflammatory agent. Agents Actions 18:407–412
70. Singh GB, Singh S, Bani S (1996) Alcoholic extract of salai-guggal ex Boswellia serrata, a
new natural source NSAID. Drugs Today 32:109–112
71. Singh S, Khajuria A, Taneja SC, Khajuria RK, Singh J, Johri RK, Qazi GN (2008) Boswellic
acids: a leukotriene inhibitor also effective through topical application in inflammatory
disorders. Phytomedicine 15:400–407
13 Boswellic Acids and Their Role in Chronic Inflammatory Diseases 327
Keywords Autoimmune Cancer Inflammation Neurodegenerative NF-κB
Withaferin A Withanolide
Abbreviations
5XFAD 5 FAD mutations carried on APP and PS1 transgenes
AChE Acetylcholinesterase
AD Alzheimer’s disease
ADAM10 Adisintegrin and metalloproteinase domain-containing protein 10, or
α-secretase
AP-1 Activator protein 1
APP Amyloid precursor protein
BACE APP cleaving enzyme 1 or β-secretase
BChE Butyrylcholinesterase
Bfl-1/A1 Bcl-2-related protein A1
C/EBPα CCAAT/enhancer-binding proteinα
CCR7 Chemokine (C–C motif) receptor 7
cFLIP C-FADD-like IL-1β-converting enzyme–inhibitory proteins
CNS Central nervous system
COX Cyclooxygenase
CSC Cancer stem cell
EGF Epidermal growth factor
EGFR EGF receptor
ERK Extracellular signal-regulated kinase
FDA Food and Drug Administration
GABA Gamma-aminobutyric acid
GMP Good manufacturing process
HD Huntington’s disease
HIF-1 Hypoxia inducible factor-1
HMGB1 High mobility group box 1
hnRNP-K Heterogeneous nuclear ribonucleoprotein
KHPA Hypothalamic–pituitary–adrenal
Hsp Heat shock protein
HTT Mutant Huntingtin
IAP1 Inhibitor of apoptosis protein-1
IBD Inflammatory bowel disease
ICAM Intercellular adhesion molecule
IFN Interferon
IKK I kappa B kinase
IL Interleukin
iNOS Inducible nitric oxide synthase
JAK Janus kinase
JNK1 c-Jun N-terminal protein kinase
Keap1 Kelch like ECH-associated protein-1
LPS Lipopolysaccharide
14 Natural Withanolides in the Treatment of Chronic Diseases 331
LTB4 Leukotriene B4
MAPK Mitogen-activated protein kinase
MCE Mitotic clonal expansion,
MCP-1 Monocyte chemoattractant protein-1
MMPs Matrix metalloproteinases
MMTV-neu Mouse mammary tumor virus-neu
MPTP 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
MS Mass spectrometry
mTOR Mechanistic target of rapamycin
MUC-1 Mucin 1
NF-κB Nuclear factor kappa B
NO Nitric oxide
Nrf2 Nuclear factor erythroid 2-related factor 2
OCD Obsessive-compulsive disorder
PBMC Peripheral blood mononuclear cells
PD Parkinson’s disease
PDGF Platelet-derived growth factor
PGE2 Prostaglandin E2
PI3K Phosphatidylinositol-3-kinase
PPARs Peroxisome proliferator-activated receptors
QR Quinone reductase
RA Rheumatoid arthritis
ROS Reactive oxygen species
RTKs Receptor tyrosine kinases
SAR Structural–activity relationship
SFMC Synovial fluid mononuclear cells
SOD Super oxide dismutase
STAT Signal transducer and activator of transcription
TAK1 Transforming growth factor-β-activating kinase
TGF-β Transforming growth factor beta
Th T-helper
TLRs Toll-like receptors
TNBS Trinitrobenzyl sulfonic acid
TNF Tumor necrosis factor
TRAIL Tumor necrosis factor-related apoptosis-inducing ligand
TWIST Twist family BHLH transcription factor
UPLC Ultra-performance liquid chromatography
VEGF Vascular endothelial growth factor
WA Withaferin A
WS Withania somnifera
332 P.T. White et al.
14.1 Introduction
Me
Me Me Me
Me 25
O
Me H Me H 24
26
OH O OH O O 23 O
Me O O O 22
Me
21
OH O Me H HO Me OH 20
OH H O
H Me O HO Me OH
H O Me H
OH Me O
H H Me H
O H H
H H
OH Cl
OH
Jaborosalactone 7 Salpichrolide G Jaborosalactone 37 Jaborosalactone 43
(Ring-A aromatic withanolides) (Ring-D aromatic withanolides) (Sativolides) (Spiranoid δ-lactones)
Me O Me
Me 25 26 Me Me
Me 24
23 25
24 O O H
HO H HO 21
22 26
O
Me H 23 O O
O Me Me
20 H H O
21
O O O
H O OH 22 Me
H O Me 20
O O Me H OH H H O
H OH O Me H
Me Me H
H H H H
H H H H
OH O
OH Cl OH
Subtrifloralactone E Tubonolide A Withametelinone Withaphysalin N
(Subtriflora-δ-lactones) (Withajardins) (Withametelins) (Withaphysalins)
Me O HO Me
26
Me 25 O
Me O O
OAc 23 24 Me
H H AcO Me 23
H
Me H O
O AcO 26
22 Me
H OH Me H H 24 O
O HO Me OH
O H Me 25
Me H O O H H OAc Me OH O
Me H
OH
OH H H
O H H
OH O
O
Subtrifloralactone K Taccalonolide A Jaborosalactol 24
(Subtriflora-γ-lactones) (Taccalonolide-γ-lactones) (Trechonolides)
Fig. 14.2 Withanolides with a γ-lactone ring and unclassified structural type
modified skeleton (e.g., physalin C and withametelinone) [1, 2, 4]. The occurrence
of unmodified withanolides is more common in nature with approximately 580 of
these naturally occurring withanolides reported in the family Solanaceae alone [1–
3]. Structurally more complex withanolides with a modified skeleton both in the
steroid nucleus and the side chain could possibly result from the biogenetic
transformations of unmodified withanolides [3, 5].
Withaferin A (WA), an archetype of this class was discovered from Withania
somnifera (WS) or Ashwagandha in 1965 [6]. In the past 50 years, approximately
900 withanolides falling into 24 diverse structural types have been discovered [1,
3]. Withanolides are mainly distributed in various genera of Solanaceae, which
includes Acnistus, Aureliana, Brachistus, Browallia, Datura, Deprea, Discopodium,
Dunalia, Exodeconus, Hyoscyamus, Iochroma, Jaborosa, Larnax, Lycium,
Mandragora, Nicandra, Physalis, Salpichroa, Saracha, Solanum, Trechonaetes,
Tubocapsicum, Vassobia, Withania, and Witheringia [1–3, 7–9]. A minor popu-
lation of withanolides has been isolated from other plant sources such as
Dioscoreaceae, Fabaceae, Labiatae, Lamiaceae, Leguminosae, Myrtaceae, and
Taccaceae [1–3] and interestingly from marine sources of Alcyoniidae family
[10–12].
334 P.T. White et al.
Me H
OAc O-Glc
Me O
OAc O Tacca
Me H O TNF -induced
Chantriolide A plantaginea Inflammation (29)
O H H NF- B
(Dioscoreaceae)*
OH O
Chantriolide A
Glc = β -D-Glucopyranosyl
Me
OH
OH
Me H
Me
Me O H
O OH
OH
Me H
H H superoxide anion
Cilistol G Solanum and elastase
Cilistol G, Capsisteroids Me
Inflammation
Me OAc OH capsicoides release by (9)
A and E Me H
OH and cancer
Me H
OH Me (Solanaceae) human
Me Me O H
Me O H O OH neutrophils
O OH Me H OH
OH
Me H
H H
H H
OH
Capsisteroid A Capsisteroid E
Me
HO H Me
Me
Me O Withania PPAR , Obesity and
O OH O
Me H
Coagulin L coagulans C/EBP , and metabolic (65)
H OH
Glc-O
(Solanaceae) MCE syndrome
Coagulin L
Glc = β-D-Glucopyranosyl
OH OH
Me H Me Me H Me
Me Me
O O
OH H O OH H O
Me H Me H
H H H H
Daturafolisides A and B, Glc-O OH Glc-O OMe
Daturafoliside A Daturafoliside B Datura LPS-induced NO
Baimantuoluoside B, and
Me Me metel(Solanaceae production in Inflammation (27)
12-
Me H Me H ) macrophages
OH O-Glc OH
Deoxywithastramonolide Me O Me O
O H O O H O
Me H Me H
H H H H
OH O OH O
Baimantuoluoside B 12-Deoxywithastramonolide
Glc = β-D-Glucopyranosyl
H
Me
OH
Me Increases
H O O
Me H
Spinal cord
Denosomin Synthetic vimentin, (66)
H H injury
HO
neuroprotective
Denosomin
Me
Me H
Me
H OH
Me Me
OH H O O
2,3-Dihydrowithaferin A Me O
H O O
Me H
O
Me H
Withania
and 4-(2,2-Dimethyl-3- H H Inflammation
H H somnifera(Solana COX-2 (30)
oxocyclopropoxy)-2,3- O
O
O
and cancer
O ceae)
dihydrowithaferin A OH
Me Me
4-(2,2-Dimethyl-3-oxocyclopropoxy)-
2,3-Dihydrowithaferin A
2,3-dihydrowithaferin A
Me
Me H Me
OH
6 ,7 -Epoxy-1-oxo- Me O
O OH O Discopodium
5 ,12 ,17 - Me H LTB4 and COX- Inflammation
H H
penninervium(So (26)
trihydroxywitha-2,24- 2 and cancer
OH O
lanaceae)
dienolide 6α,7α-Epoxy-1-oxo-5α,12α,17α-
trihydroxywitha-2,24-dienolide
Me
HO H Me
Me
Me O
O OH O Physalis
4 -Hydroxywithanolide Me H NF- B and Diabetes and
pruinosa (67)
E H OH MCP-1 obesity
O
(Solanaceae)
OH
4β-Hydroxywithanolide E
336 P.T. White et al.
Formalin-
Me
HO H
induced arthritis
Me Me
Me O
Withania model and cotton
3 -Hydroxy-2,3- O OH O
Me H coagulans pellet granuloma Inflammation (68)
dihydrowithanolide F H OH (Solanaceae) method for sub-
HO
3β-Hydroxy-2,3-dihydrowithanolide F acute
inflammation
Me Me
Me H Me Me H Me
AcO
Me
O O
H O H O
Me H Me H
H H H H LPS-induced
O O Paraminabea
Minabeolides-1, -2, -4, Minabeolide 1 Minabeolide 2 iNOS and COX- Inflammation
acronocephala(A (10)
and -5 Me H Me Me H Me
2 proteins in and cancer
AcO lcyoniidae)
Me
O O macrophages
H O H O
Me H Me H
H H H H
O O
Minabeolide 4 Minabeolide 5
Me Me
Me H Me Me H Me
AcO
OHC
O O
H O H O
Me H Me H
H H H H
O O Paraminabea LPS-induced
Paraminabeolide A Paraminabeolide B Inflammation
Paraminabeolides A–D acronocephala(A iNOS protein in (10)
Me OH Me OH and cancer
AcO H Me H Me lcyoniidae) macrophages
Me
H H
Me H O Me Me H O Me
H H O H H O
O O
Paraminabeolide C Paraminabeolide D
Me H Me H
O O O O
O O IKK , NF- B,
H O H O
O O H
Physalis
Physalins A and O, and Me HO O Me HO O and LPS-induced
Me Me Me alkekengi Inflammation (64)
Isophysalin A H HOH O H HOH O NO production
OH OH (Solanaceae)
Isophysalin A Physalin O
in macrophages
Me H Me H
O O O
O O O
O H O
H
O O
Me HO O Me HO O H
Me Me
H OH O H O
H H
O
Physalin C Physalin B
Me H Me H
O O O O
O O
H O H O
O O
Me HO O Me HO O H
Me Me
H HOH O H H O
O
OH OH
Physalis
Physalin A Physalin N
Physalins C, B, A, N, F alkekengi and
Me H
Me TNF -induced Inflammation
and Withanolides E, F, O O HO H Me
Physalis (59)
O Me NF- B and cancer
H O
G O Me O peruviana
Me HO O H O OH O
Me Me H (Solanaceae)
H H O
O H OH
O
O
Physalin F Withanolide E
Me Me
HO H Me HO H Me
Me Me
Me O Me O
O OH O O H O
Me H Me H
H OH H OH
Withanolide F Withanolide G
14 Natural Withanolides in the Treatment of Chronic Diseases 337
Me
Me H Inflammation
OAc O-Glc
Me O
OH O Tacca and metabolic
Me H O
Plantagiolide I AcO plantaginea PPARs disorders such (29)
H H
Cl
OH O
(Dioscoreaceae)* as diabetes
Plantagiolide I and obesity
Glc = β-D-Glucopyranosyl
Me
H
Me
OH
Me
H O O
OH
Me H
H H
HO
Sominone
Me
Withania Dementia and
Sominone and Induces neurite (39,
Me
H somnifera(Solana Alzheimer's
Withanoside IV OH outgrowth 40)
Me ceae) disease
H O O
OH
OH Me H
HO
O H H
HO
O
OH O O
OH
HO
HO
Withanoside IV
Me
H
Me Me
Me
O O O Tubocapsicum CCR7
O
Me H
Tubocapsanolide A anomalum expression, IKK, Cancer (69)
H H
O
(Solanaceae) and NF- B
OH
TubocapsanolideA
Me
H
Me Me
Me O Tubocapsicum Hsp90-Hsp70
O O
Me H OH (62,
Tubocapsenolide A anomalum chaperone Cancer
H 70)
(Solanaceae) complex
O
OH
Tubocapsenolide A
Me Me
O O
H H
Me Me Me Me
Me Me
H O OH H O OH
O O
Me H OAc Me H
H H H H
O O
OH OH Physalis
Virginol A Virginol B TPA-induced ear
Virginols A–C virginiana(Solan Inflammation (28)
Me edema assay
O aceae)
H
Me Me
Me
H O OH
O
Me H OAc
H H
O
Virginol C
Me
H
Me TNF -induced Inflammation
Me
OH (54)
H O O Withania NF- B and cancer
O
Me H
Viscosalactone B somnifera(Solana
H H
HO
O
ceae) Inflammation
OH COX-2 (30)
Viscosalactone B
and cancer
Me
H
Me Me
AcO
H O O
O
Me H STAT3 and Inflammation
Withacnistin –† (71)
H H STAT5 and cancer
O
OH
Withacnistin
TNF -induced
Angio-
NF- B and
inflammatory
Ubiquitin (55)
diseases and
proteasome
cancer
pathway
NF- B and
338 P.T. White et al.
zymosan-
Inflammation
Me induced (24)
and cancer
H inflammed paw
Me
OH
Me Withania
H O O
model
O
Me H
Withaferin A somnifera(Solana NF- B, AP-1, Inflammation
H H (72)
ceae) Nrf2, and IL-6 and cancer
O
OH
Withaferin A
Inflammation
COX-2 (30)
and cancer
Inflammation (53,
STAT3
and cancer 57)
COX-2, PGE2,
Microglial
STAT1, and (73)
activation
STAT3
LPS-mediated Vascular
(51)
HMGB1 release, inflammatory
H H
OH COX-2 and
OH S
O Withania
OH Inflammation
Withanolide sulfoxide OH somnifera TNF -induced (74)
H H
and cancer
(Solanaceae) NF- B
H Me
O O
O H Me
HO
Me
H
Me
Withanolide sulfoxide
* Reference 28 had the genus Tacca assigned to family Taccaceae, which has been found in older
texts but the APG II system has incorporated this genus into the family Dioscoreaceae. † Obtained
from National Cancer Institute Developmental Therapeutics Program
possibly through IL-2 receptor binding [87, 88]. Similar to coagulins, physalins B,
F, G, and H isolated from Physalis angulata have demonstrated immunosuppres-
sive properties. Physalins B, F, and G showed inhibition of lipopolysaccharide
(LPS)-activated macrophages along with their cytokine (TNF-α, IL-6 and IL-12)
and nitric oxide (NO) production, whereas concanavalin A-induced T-cell prolif-
eration and cytokine production in a mechanism distinct from dexamethasone [89,
90]. Physalin H has demonstrated polarization to Th-2 cells with inhibition of Th-1
cytokines IL-2 and IFN-γ, increased Th-2 cytokines IL-4 and IL-10, and induction
of the heme oxygenase-1 [91].
340 P.T. White et al.
There are strong preclinical and clinical studies that demonstrate that inflam-
mation initially started by immune cell mediators may persist chronically, resulting
in ongoing stimulation of inflammatory mediators and regulatory pathways that
contribute to the pathogenesis of chronic diseases including cardiovascular, neu-
rologic, and pulmonary diseases, as well as cancer, diabetes, and obesity [92–94].
As with acute inflammation, chronic inflammation is mediated through various
signaling factors, which include proinflammatory cytokines such as TNFα, IL-1,
IL-6, IL-8, IL-12, NO, adhesion molecules, and chemokines [95]. Additionally,
transcription factors that regulate the expression of inflammatory mediators such as
NF-κB, activator protein 1 (AP-1), peroxisome proliferator-activated receptor
(PPAR)-γ, STAT3, hypoxia inducible factor-1 (HIF-1), β-catenin/Wnt, hedgehog,
and nuclear factor erythroid 2-related factor 2 (Nrf2) have been linked to chronic
diseases. The DNA-binding capacity of these transcription factors is modified by
several signaling cascades such as JAK/STAT, MAPKs, PI3K/Akt/mechanistic
target of rapamycin (mTOR), and ubiquitin proteasome system [96]. These sig-
naling pathways have a wide range of functions and show complex crosstalk
depending on the cell type and the chronic disease involved. Withanolides have
emerged recently as potential therapeutics for chronic diseases due to their unique
ability to modulate multiple signaling pathways. In this chapter, we will discuss
several different withanolides and their seemingly broad mechanism of action in
modulating key molecular pathways that connect inflammation and chronic
diseases.
The NF-κB family of transcription factors plays a prominent role in the immune
system and inflammation. In response to ligation by Toll-like receptors (TLRs) and
IL-1 receptor family members (B-cell and T-cell receptors), NF-κB regulates the
expression of several factors such as inhibitor of apoptosis protein-1 (IAP1),
inducible nitric oxide synthase (iNOS), cytokines, cyclooxygenase (COX)-2,
prostaglandins, growth factors, and effector enzymes [97–99]. NF-κB is activated
by inflammatory cytokines, stress, free radicals, radiation, growth receptors, and
TNFα leading to transcriptional regulation of several genes that are involved in
proliferation, inflammation, cellular survival, apoptosis, angiogenesis, and differ-
entiation [100–103]. The mammalian transcription NF-κB family of proteins
includes RelA (p65), RelB, NF-κB2 (p52), c-Rel, and NF-κB1 (p50). In the absence
of stimuli, inactive forms of NF-κB proteins are present in the cytoplasm due to
their interaction with several inhibitors of kappaB (IκB) proteins. Upon exposure to
stimuli, NF-κB is activated either through canonical or noncanonical pathways by
regulatory IκB kinases (IKK) such as IKKα, IKKβ, and IKKγ. In the most common
classical pathway, IKK phosphorylation leads to IκBα phosphorylation at serine 32
and serine 36, followed by phosphorylation, nuclear localization, DNA binding of
p65–p50 complex, and transcriptional activation of NF-κB responsive genes [104].
14 Natural Withanolides in the Treatment of Chronic Diseases 341
The transcription factor AP-1, which plays a key role in the inflammatory response
is implicated in several diseases such as cancer, psoriasis, inflammatory bowel
disease (IBD), rheumatoid arthritis (RA) and fibrosis [117]. The AP-1 complex
consists of homo and hetero dimers of Jun (JunD, C-Jun, and JunB) and the Fos
(FosB, C-Fos, Fra-1 and Fra-2) family of proteins [118, 119]. Cytokines,
chemokines, hormones, and growth factors as well as external stress factors are
known to activate AP-1 signaling. The AP-1 complex translocates to the nucleus in
response to stress signaling cascades, such MAPKs and c-Jun terminal kinases
[120]. This in turn leads to activation of AP-1 and regulates multiple functions such
as differentiation, transformation, proliferation, and survival [121]. The crude
ethanol extract of WS has been shown to inhibit the nuclear localization of both
AP-1 and NF-κB in LPS-activated peripheral blood mononuclear cells (PBMC) of
both normal and RA patients, as well as synovial fluid mononuclear cells (SFMC)
of RA patients. This in turn led to decreased downstream transcription target genes
such as MMPs, COX-2, and iNOS, all of which are known mediators of RA [122].
PPARγ was first discovered in adipocyte differentiation and lipid metabolism and is
one of three members in this nuclear receptor family of transcription factors [123].
The other members of the PPARs in mammals are PPARα and PPARβ/δ. The
PPARs activate several genes involved in inflammation, adipogenesis, lipid meta-
bolism, glucose metabolism, cellular differentiation, development, and tumorigen-
esis via binding of the PPAR/retinoid X receptor heterodimer to PPAR-responsive
regulatory elements [124, 125]. PPARγ plays a key role in inflammation through
modulation of proinflammatory transcription factors such as NF-κB and AP-1
[113]. Treatment of 3T3-L1 adipocytes with WA resulted in phosphorylation of
extracellular signal-regulated kinase (ERK), followed by decreased expressions of
PPARγ leading to altered levels of Bcl2 and Bax expression, induction of apoptosis,
14 Natural Withanolides in the Treatment of Chronic Diseases 345
Nuclear factor erythroid 2 related factor 2(Nrf2) is a transcription factor that reg-
ulates genes involved in redox homeostasis, inflammation, energy metabolism and
cellular growth [146]. Under normal homeostatic conditions, Nrf2 is anchored in
the cytoplasm as a complex with Kelch like ECH-associated protein-1 (Keap1),
which facilitates ubiquitin mediated proteasome degradation of Nrf2 and decreased
expression of Nrf2 target genes. However, in response to stimulation by growth
factors, electrophilic stressors, and changes in redox signal, Nrf2 ubiquitination is
disrupted and levels increase rapidly. Nrf2 translocates to the nucleus and upreg-
ulates expression of proteins involved in glutathione and thioredoxin-based
antioxidant defense, drug metabolism and efflux, and proteins associated with
heme and iron metabolism [147]. Nrf2 is engaged in crosstalk with several sig-
naling pathways that play a critical role in the pathogenesis and progression of
chronic diseases, including NF-κB, PI3K, MAPK, glycogen synthase kinase-3β,
and notch [146, 147]. Molecular docking studies have shown that both WA and
withanone interact with the amino acids Ala 69, Gln 75, and Phe 71 of Nrf2 [148].
In another study, WA induced reactive oxygen species (ROS) that activated JNK
and stabilized Nrf2 that resulted in activation of NADPH quinone oxidoreductase
and Tap73 transcriptional function leading to apoptosis of cancer cells [149]. WA
was also shown to inhibit NFκB, AP-1, and Nrf2 in adriamycin-resistant human
myelogenous erythroleukemic K562/Adr cells in a dose-dependent manner [72].
Moreover, compared to other tested natural products such as quercetin, only WA
overcomes attenuated caspase activation and blocking of apoptosis in K562/Adr
cells [72].
Under normal oxygen conditions, the HIF-1 α protein is synthesized at a high rate
and rendered transcriptionally inactive due to immediate hydroxylation-dependent
proteasome/ubiquitin degradation by the VHL E3 ligase. However, when hypoxia
is induced through impaired cellular oxygen balance, hydroxylase activity is
downregulated, HIF-α protein is stabilized, and HIF-1 is activated [150].
Transcriptional activation of HIF-1 upregulates several genes that control glycolytic
metabolism, angiogenesis, invasion, metastasis, and cell survival, such as VEGF,
MMPs, stromal cell-derived factor-1, e-cadherin, chemokine receptor 4, EGF, and
transforming growth factor beta (TGF-β) 3 [151–155]. Crosstalk between NF-κB
and HIF pathways has been shown to be associated with several chronic inflam-
matory diseases such as cancer, RA, asthma, and chronic obstructive pulmonary
diseases [156]. In solid tumors, the availability of oxygen within the tumor
decreases as distance from blood vessels increases resulting in the creation of
hypoxic regions [157]. This is known to be responsible in part for therapy resistance
14 Natural Withanolides in the Treatment of Chronic Diseases 347
and metastatic spread [158]. Although, no study thus far directly demonstrates
inhibition of HIF-1 transcriptional activation by withanolides, a few note down-
regulation of migration-promoting HIF-mediated genes such as VEGF, heteroge-
neous nuclear ribonucleoprotein K (hnRNP-K) and MMPs, which lead to restriction
of angiogenesis and metastasis [159].
There have been several studies indicating that withanolides have a chrondropro-
tective aspect. WS extract has been shown in animal models to significantly reduce
the effects of collagenase on bovine Achilles tendons with even a suggestion of
collagen stabilization [167]. This has also been demonstrated with inhibition of the
gelatinase activity of collagenase type 2 enzyme in vitro [168]. Several
collagen-induced arthritic rat models have noted significant amelioration of paw
and ankle arthritis, oxidative stress, degradation of cartilage, and improvements in
functional recovery and radiological score [24, 169–171]. Aqueous extracts of WS
have demonstrated a significant dose-dependent reduction in adjuvant-induced hind
foot pad thickness, reductions in immune complement activity [172], as well as
reduced arthritic index, autoantibodies, and CRP levels with results comparable to
methotrexate treatment. These rats similarly demonstrate reductions in oxidative
stress through decreased lipid peroxidation, glutathione-S-transferase activity, and
14 Natural Withanolides in the Treatment of Chronic Diseases 349
The discovery that most withanolides have cytotoxic capabilities against a wide
range of cancers has initiated a large breadth of research into the cytotoxic
mechanisms and potential therapeutic benefits of withanolides for cancer treatment
and prevention. As previously described and depicted in Fig. 14.3, withanolides
inhibit multiple aspects of inflammatory pathways. While these inflammatory
pathways have complex interactions between one another, they also interact with
proliferative and oncogenic pathways. Several key mechanisms of cytotoxicity
from withanolides have been described and these include: (a) induction of oxidative
stress, (b) inhibition of proteasome mediated ubiquitin degradation of IκBα (leading
to inhibition of NF-κB and its downstream effectors AP-1 and Nrf-2), (c) inhibition
of transcription factor STAT3, (d) inhibition of Hsp90 (by blocking interaction of
the Cdc37 co-chaperone with Hsp90) with resultant Hsp90 client inhibition in the
PI3K/Akt and MAPK pathways, (e) dysregulation of cytoskeletal and structural
proteins, and (f) angiogenesis inhibition though HIF-1 [1, 137, 182, 183].
Withanolides with significant activity and identified mechanisms are depicted in
Table 14.1, and their cytotoxic characteristics have been demonstrated in multiple
cancer types, including breast, ovarian, colon, head and neck, renal, prostate,
pancreatic, thyroid, glioblastoma, and hematologic cancers such as lymphoma,
leukemia, and multiple myeloma [1, 70, 136, 182–186].
lymphomas [199], ovarian cancer [200, 201], prostate cancer [202, 203], soft tissue
sarcoma [204], cervical carcinoma [205], melanoma, and mesothelioma [206].
These exciting results all point toward the incredible potential of these natural
products for future clinical treatment regimens.
more recently been shown to have key anti-inflammatory interactions within the
immune system through suppression of T-cells and macrophages, and inhibition of
NF-κB on lymphocytes [242]. The GABA-mimetic activity of WS root extract has
been shown for several decades [243] with a recent study identifying a 27 times
greater affinity of WS for the highly sensitive GABAρ1 receptors compared to
GABAA receptors, though WA and withanolide A were not the active
GABA-mimetics [244]. These GABA-mimetic and adaptogenic effects likely play a
significant role in the long-standing history of using Ashwagandha for the treatment
of anxiety and neurobehavioral disorders, though the vast majority of animal and
human studies have been performed in the last several decades.
Most animal studies evaluating neurologic disorders have focused on the effects of
WS extract using models of either chronic stress or withdrawal to induce anxiety
and depression. Using a chronic, unpredictable, mild footshock to create a chronic
stress model in rats, treatment with aqueous ethanol WS root extract significantly
attenuated chronic, stress-induced abnormalities with improvements in biochemical
imbalances (elevated blood glucose and corticosteroid levels), reduced number and
severity of gastric ulcers, improved behavioral depression and sexual responses,
improved cognitive memory function, and rescued immunosuppression in macro-
phage activity and immunologic pedal edema [245]. In depression and anxiety
rodent models using chronic stress, isolation, sleep deprivation, WS extract treat-
ment demonstrated significant antidepressant effects, and potentiated conventional
antidepressants (tricyclic antidepressant imipramine and selective
serotonin-reuptake inhibitor fluoxetine) measured by reduced immobility time in
the forced open swim test (“behavioral despair/learned helplessness”), and an
anxiolytic effect similar to benzodiazepines (lorazepam and diazepam) in the ele-
vated plus maze, the social interaction test, and the feeding latency test [246–250].
In an OCD rodent model of marble-burying behavior, intraperitoneal injections
of both methanolic and aqueous WS root extracts 30 min prior to evaluation
resulted in significant dose-dependent reductions in marble-burying behavior sim-
ilar to standard fluoxetine alone, and a synergistic effect in combination with
fluoxetine [251]. These in vivo studies demonstrate a consistent amelioration of
stress, anxiety, depressant, and OCD behaviors with corresponding corrections in
biochemical abnormalities, however, the modulation of inflammatory pathways in
these diseases requires further characterization.
14 Natural Withanolides in the Treatment of Chronic Diseases 359
alterations in inflammatory cytokines [260]. The results from this study will help
provide valuable insight into the effect of WS on neuroinflammation and
schizophrenia symptoms.
14.7 Conclusions
References
1. Chen L-X, He H, Qiu F (2011) Natural withanolides: an overview. Nat Prod Rep 28(4):705–
740
2. Misico R, Nicotra V, Oberti J, Barboza G, Gil R, Burton G (2011) Withanolides and related
steroids. In: Kinghorn AD, Falk H, Kobayashi J (eds) Progress in the chemistry of organic
natural products. Fortschritte der Chemie organischer Naturstoffe/Progress in the chemistry
of organic natural products, vol 94. Springer, Vienna, pp 127–229
3. Zhang H, Cao C-M, Gallagher RJ, Timmermann BN (2014) Antiproliferative withanolides
from several solanaceous species. Nat Prod Res 28(22):1941–1951
4. Glotter E (1991) Withanolides and related ergostane-type steroids. Nat Prod Rep 8(4):415–
440
5. Misico RI, Song LL, Veleiro AS, Cirigliano AM, Tettamanzi MC, Burton G et al (2002)
Induction of quinone reductase by withanolides. J Nat Prod 65(5):677–680
6. Lavie D, Glotter E, Shvo Y (1965) Constituents of Withania somnifera Dun. Part IV. The
structure of withaferin A. J Chem Soc (1): 7517–7531
14 Natural Withanolides in the Treatment of Chronic Diseases 361
27. Yang B-Y, Guo R, Li T, Wu J-J, Zhang J, Liu Y et al (2014) New anti-inflammatory
withanolides from the leaves of Datura metel L. Steroids 87:26–34
28. Maldonado E, Amador S, Martínez M, Pérez-Castorena AL (2010) Virginols A–C, three new
withanolides from Physalis virginiana. Steroids 75(4–5):346–349
29. Quang TH, Ngan NTT, Minh CV, Kiem PV, Yen PH, Tai BH et al (2012) Plantagiolides I
and J, two new withanolide glucosides from Tacca plantaginea with nuclear factor-kappaB
inhibitory and peroxisome proliferator-activated receptor transactivational activities. Chem
Pharm Bull 60(12):1494–1501
30. Jayaprakasam B, Nair MG (2003) Cyclooxygenase-2 enzyme inhibitory withanolides from
Withania somnifera leaves. Tetrahedron 59(6):841–849
31. Qiu L, Zhao F, Jiang Z-H, Chen L-X, Zhao Q, Liu H-X et al (2008) Steroids and flavonoids
from Physalis alkekengi var. franchetii and their inhibitory effects on nitric oxide production.
J Nat Prod 71(4):642–646
32. Malik F, Singh J, Khajuria A, Suri KA, Satti NK, Singh S et al (2007) A standardized root
extract of Withania somnifera and its major constituent withanolide-A elicit humoral and
cell-mediated immune responses by up regulation of Th1-dominant polarization in BALB/c
mice. Life Sci 80(16):1525–1538
33. Habtemariam S (1997) Cytotoxicity and immunosuppressive activity of withanolides from
Discopodium penninervium. Planta Med 63(01):15–17
34. Kour K, Pandey A, Suri KA, Satti NK, Gupta KK, Bani S (2009) Restoration of
stress-induced altered T cell function and corresponding cytokines patterns by
Withanolide A. Int Immunopharmacol 9(10):1137–1144
35. Abou-Douh AM (2002) New withanolides and other constituents from the fruit of Withania
somnifera. Arch Pharm 335(6):267–276
36. Bravo BJA, Sauvain M, Gimenez TA, Balanza E, Serani L, Laprévote O et al (2001)
Trypanocidal withanolides and withanolide glycosides from Dunalia brachyacantha. J Nat
Prod 64(6):720–725
37. Choudhary MI, Yousuf S, Samreen AS, Atta UR (2007) New leishmanicidal physalins from
Physalis minima. Nat Prod Res 21(10):877–883
38. Bhattacharya SK, Satyan KS, Ghosal S (1997) Antioxidant activity of glycowithanolides
from Withania somnifera. Indian J Exp Biol 35(3):236–239
39. Kuboyama T, Tohda C, Komatsu K (2006) Withanoside IV and its active metabolite,
sominone, attenuate Aβ(25–35)-induced neurodegeneration. Eur J Neurosci 23(6):1417–
1426
40. Joyashiki E, Matsuya Y, Tohda C (2011) Sominone Improves Memory Impairments and
Increases axonal density in Alzheimer’s Disease Model Mice, 5XFAD. Int J Neurosci 121
(4):181–190
41. Baitharu I, Jain V, Deep SN, Shroff S, Sahu JK, Naik PK et al (2014) Withanolide A prevents
neurodegeneration by modulating hippocampal glutathione biosynthesis during hypoxia.
PLoS ONE 9(10):e105311
42. Devi PU (1996) Withaferin A: a new radiosensitizer from the Indian medicinal plant
Withania somnifera. Int J Radiat Biol 69(2):193–197
43. Devi PU, Kamath R (2003) Radiosensitizing effect of withaferin A combined with
hyperthermia on mouse fibrosarcoma and melanoma. J Radiat Res 44(1):1–6
44. Budhiraja RD, Krishan P, Sudhir S (2000) Biological activity of withanolides. J Sci Ind Res
59(11):904–911
45. Mareggiani G, Picollo MI, Zerba E, Burton G, Tettamanzi MC, Benedetti-Doctorovich MOV
et al (2000) Antifeedant Activity of Withanolides from Salpichroa origanifolia on Musca
domestica. J Nat Prod 63(8):1113–1116
46. Mandal C, Dutta A, Mallick A, Chandra S, Misra L, Sangwan R et al (2008) Withaferin A
induces apoptosis by activating p38 mitogen-activated protein kinase signaling cascade in
leukemic cells of lymphoid and myeloid origin through mitochondrial death cascade.
Apoptosis 13(12):1450–1464
14 Natural Withanolides in the Treatment of Chronic Diseases 363
47. Khan ZA, Ghosh AR (2010) Possible nitric oxide modulation in protective effects of
withaferin A against stress induced neurobehavioural changes. J Med Plants Res 4(6):490–
495
48. Machin RP, Veleiro AS, Nicotra VE, Oberti JC, Padrón JM (2010) Antiproliferative activity
of withanolides against human breast cancer cell lines. J Nat Prod 73(5):966–968
49. Koduru S, Kumar R, Srinivasan S, Evers MB, Damodaran C (2010) Notch-1 inhibition by
Withaferin-A: a therapeutic target against colon carcinogenesis. Mol Cancer Ther 9(1):202–
210
50. Rah B, Amin H, Yousuf K, Khan S, Jamwal G, Mukherjee D et al (2012) A novel MMP-2
inhibitor 3-azidowithaferin A (3-azidoWA) abrogates cancer cell invasion and angiogenesis
by modulating extracellular par-4. PLoS ONE 7(9):e44039
51. Lee W, Kim TH, Ku S-K, K-j Min, Lee H-S, Kwon TK et al (2012) Barrier protective effects
of withaferin A in HMGB1-induced inflammatory responses in both cellular and animal
models. Toxicol Appl Pharmacol 262(1):91–98
52. Bargagna-Mohan P, Ravindranath PP, Mohan R (2006) Small molecule anti-angiogenic
probes of the ubiquitin proteasome pathway: Potential application to choroidal
neovascularization. Invest Ophthalmol Vis Sci 47(9):4138–4145
53. Choi BY, Kim B-W (2015) Withaferin-A inhibits colon cancer cell growth by blocking
STAT3 transcriptional activity. J Cancer Prevent 20(3):185–192
54. Ichikawa H, Takada Y, Shishodia S, Jayaprakasam B, Nair MG, Aggarwal BB (2006)
Withanolides potentiate apoptosis, inhibit invasion, and abolish osteoclastogenesis through
suppression of nuclear factor-κB (NF-κB) activation and NF-κB-regulated gene expression.
Mol Cancer Ther 5(6):1434–1445
55. Mohan R, Hammers H, Bargagna-Mohan P, Zhan X, Herbstritt C, Ruiz A et al (2004)
Withaferin A is a potent inhibitor of angiogenesis. Angiogenesis 7(2):115–122
56. Ndlovu MN, Van Lint C, Van Wesemael K, Callebert P, Chalbos D, Haegeman G et al
(2009) Hyperactivated NF-κB and AP-1 transcription factors promote highly accessible
chromatin and constitutive transcription across the interleukin-6 gene promoter in metastatic
breast cancer cells. Mol Cell Biol 29(20):5488–5504
57. Yco LP, Mocz G, Opoku-Ansah J, Bachmann AS (2014) Withaferin A Inhibits STAT3 and
induces tumor cell death in neuroblastoma and multiple myeloma. Biochem Insights 7:1–13
58. Ishiguro M, Kajikawa A, Haruyama T, Morisaki M, Ikekawa N (1974) Synthetic studies of
withanolide I synthesis of AB ring moiety of withaferin A. Tetrahedron Lett 15(15):1421–
1424
59. Ozawa M, Morita M, Hirai G, Tamura S, Kawai M, Tsuchiya A et al (2013) Contribution of
cage-shaped structure of physalins to their mode of action in inhibition of NF-κB activation.
ACS Med Chem Lett 4(8):730–735
60. Wijeratne EMK, Xu Y-M, Scherz-Shouval R, Marron MT, Rocha DD, Liu MX et al (2014)
Structure–activity relationships for withanolides as inducers of the cellular heat-shock
response. J Med Chem 57(7):2851–2863
61. Damu AG, Kuo P-C, Su C-R, Kuo T-H, Chen T-H, Bastow KF et al (2007) Isolation,
structures, and structure-cytotoxic activity relationships of withanolides and physalins from
Physalis angulata. J Nat Prod 70(7):1146–1152
62. Wang H-C, Tsai Y-L, Wu Y-C, Chang F-R, Liu M-H, Chen W-Y et al (2012)
Withanolides-induced breast cancer cell death is correlated with their ability to inhibit heat
protein 90. PLoS ONE 7(5):e37764
63. Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B et al (2010) Withaferin A targets heat
shock protein 90 in pancreatic cancer cells. Biochem Pharmacol 79(4):542–551
64. Ji L, Yuan Y, Luo L, Chen Z, Ma X, Ma Z et al (2012) Physalins with anti-inflammatory
activity are present in Physalis alkekengi var. franchetii and can function as Michael reaction
acceptors. Steroids 77(5):441–447
65. Beg M, Chauhan P, Varshney S, Shankar K, Rajan S, Saini D et al (2014) A withanolide
coagulin-L inhibits adipogenesis modulating Wnt/β-catenin pathway and cell cycle in mitotic
clonal expansion. Phytomedicine 21(4):406–414
364 P.T. White et al.
84. Davis L, Kuttan G (2002) Effect of Withania somnifera on cell mediated immune responses
in mice. J Exp Clin Cancer Res. 21(4):585–590
85. Khan B, Ahmad SF, Bani S, Kaul A, Suri KA, Satti NK et al (2006) Augmentation and
proliferation of T lymphocytes and Th-1 cytokines by Withania somnifera in stressed mice.
Int Immunopharmacol 6(9):1394–1403
86. Bani S, Gautam M, Sheikh FA, Khan B, Satti NK, Suri KA et al (2006) Selective Th1
up-regulating activity of Withania somnifera aqueous extract in an experimental system
using flow cytometry. J Ethnopharmacol 107(1):107–115
87. Mesaik MA, Zaheer Ul H, Murad S, Ismail Z, Abdullah NR, Gill HK et al (2006) Biological
and molecular docking studies on coagulin-H: Human IL-2 novel natural inhibitor. Mol
Immunol 43(11):1855–1863
88. Huang C-F, Ma L, Sun L-J, Ali M, Arfan M, Liu J-W et al (2009) Immunosuppressive
withanolides from withania coagulans. Chem Biodivers 6(9):1415–1426
89. Soares MB, Bellintani MC, Ribeiro IM, Tomassini TC, dos Santos RR (2003) Inhibition of
macrophage activation and lipopolysaccaride-induced death by seco-steroids purified from
Physalis angulata L. Eur J Pharmacol 459(1):107–112
90. Soares MBP, Brustolim D, Santos LA, Bellintani MC, Paiva FP, Ribeiro YM et al (2006)
Physalins B, F and G, seco-steroids purified from Physalis angulata L., inhibit lymphocyte
function and allogeneic transplant rejection. Int Immunopharmacol 6(3):408–414
91. Yu Y, Sun L, Ma L, Li J, Hu L, Liu J (2010) Investigation of the immunosuppressive activity
of Physalin H on T lymphocytes. Int Immunopharmacol 10(3):290–297
92. Aggarwal BB, Vijayalekshmi RV, Sung B (2009) Targeting inflammatory pathways for
prevention and therapy of cancer: short-term friend, long-term foe. Clin Cancer Res 15
(2):425–430
93. Costa G, Francisco V, Lopes MC, Cruz MT, Batista MT (2012) intracellular signaling
pathways modulated by phenolic compounds: application for new anti-inflammatory drugs
discovery. Curr Med Chem 19(18):2876–2900
94. Libby P (2007) Inflammatory mechanisms: the molecular basis of inflammation and disease.
Nutr Rev 65(12 Pt 2):S140–S146
95. Santangelo C, Vari R, Scazzocchio B, Di Benedetto R, Filesi C, Masella R (2007)
Polyphenols, intracellular signalling and inflammation. Ann Ist Super Sanita 43(4):394–405
96. O’Neill LA (2006) Targeting signal transduction as a strategy to treat inflammatory diseases.
Nat Rev Drug Discov 5(7):549–563
97. Ghosh DM, George BS, Bhatia A, Sidhu OP (2014) Characterization of Withania somnifera
leaf transcriptome and expression analysis of pathogenesis-related genes during salicylic acid
signaling. PLoS ONE 9(4):e94803
98. Li Q, Verma IM (2002) NF-kappaB regulation in the immune system. Nat Rev Immunol 2
(10):725–734
99. Bonizzi G, Karin M (2004) The two NF-kappaB activation pathways and their role in innate
and adaptive immunity. Trends Immunol 25(6):280–288
100. Brasier AR (2006) The NF-kappaB regulatory network. Cardiovasc Toxicol 6(2):111–130
101. Gilmore TD (2006) Introduction to NF-kappaB: players, pathways, perspectives. Oncogene
25(51):6680–6684
102. Perkins ND (2007) Integrating cell-signalling pathways with NF-kappaB and IKK function.
Nat Rev Mol Cell Biol 8(1):49–62
103. Tian B, Brasier AR (2003) Identification of a nuclear factor kappa B-dependent gene
network. Recent Prog Horm Res 58:95–130
104. Karin M (2009) NF-kappaB as a critical link between inflammation and cancer. Cold Spring
Harb Perspect Biol 1(5):a000141
105. Darnell JE Jr (1997) STATs and gene regulation. Science 277(5332):1630–1635
106. Darnell JE Jr (2002) Transcription factors as targets for cancer therapy. Nat Rev Cancer 2
(10):740–749
107. Yu H, Pardoll D, Jove R (2009) STATs in cancer inflammation and immunity: a leading role
for STAT3. Nat Rev Cancer 9(11):798–809
366 P.T. White et al.
108. Yu H, Jove R (2004) The STATs of cancer—new molecular targets come of age. Nat Rev
Cancer 4(2):97–105
109. Takeda K, Noguchi K, Shi W, Tanaka T, Matsumoto M, Yoshida N et al (1997) Targeted
disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad
Sci USA 94(8):3801–3804
110. Cheng GZ, Zhang WZ, Sun M, Wang Q, Coppola D, Mansour M et al (2008) Twist is
transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function.
J Biol Chem 283(21):14665–14673
111. Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J et al (2002) Constitutive Stat3
activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 21(13):2000–
2008
112. Ahmad R, Rajabi H, Kosugi M, Joshi MD, Alam M, Vasir B et al (2011) MUC1-C
oncoprotein promotes STAT3 activation in an autoinductive regulatory loop. Sci Signal 4
(160):ra9
113. Kiuchi N, Nakajima K, Ichiba M, Fukada T, Narimatsu M, Mizuno K et al (1999) STAT3 is
required for the gp130-mediated full activation of the c-myc gene. J Exp Med 189(1):63–73
114. Masuda M, Suzui M, Yasumatu R, Nakashima T, Kuratomi Y, Azuma K et al (2002)
Constitutive activation of signal transducers and activators of transcription 3 correlates with
cyclin D1 overexpression and may provide a novel prognostic marker in head and neck
squamous cell carcinoma. Cancer Res 62(12):3351–3355
115. Lee J, Hahm ER, Singh SV (2010) Withaferin A inhibits activation of signal transducer and
activator of transcription 3 in human breast cancer cells. Carcinogenesis 31(11):1991–1998
116. Um HJ, Min K-J, Kim DE, Kwon TK (2012) Withaferin A inhibits JAK/STAT3 signaling
and induces apoptosis of human renal carcinoma Caki cells. Biochem Biophys Res Commun
427(1):24–29
117. Schonthaler HB, Guinea-Viniegra J, Wagner EF (2011) Targeting inflammation by
modulating the Jun/AP-1 pathway. Ann Rheum Dis 70(Suppl 1):i109–i112
118. Zenz R, Wagner EF (2006) Jun signalling in the epidermis: From developmental defects to
psoriasis and skin tumors. Int J Biochem Cell Biol 38(7):1043–1049
119. Hess J, Angel P, Schorpp-Kistner M (2004) AP-1 subunits: quarrel and harmony among
siblings. J Cell Sci 117(Pt 25):5965–5973
120. Wagner EF, Nebreda AR (2009) Signal integration by JNK and p38 MAPK pathways in
cancer development. Nat Rev Cancer 9(8):537–549
121. Eferl R, Wagner EF (2003) AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer 3
(11):859–868
122. Singh D, Aggarwal A, Maurya R, Naik S (2007) Withania somnifera inhibits NF-kappaB and
AP-1 transcription factors in human peripheral blood and synovial fluid mononuclear cells.
Phytother Res 21(10):905–913
123. Pratt WB, Toft DO (2003) Regulation of signaling protein function and trafficking by the
hsp90/hsp70-based chaperone machinery. Exp Biol Med (Maywood) 228(2):111–133
124. Sawai A, Chandarlapaty S, Greulich H, Gonen M, Ye Q, Arteaga CL et al (2008) Inhibition
of Hsp90 down-regulates mutant epidermal growth factor receptor (EGFR) expression and
sensitizes EGFR mutant tumors to paclitaxel. Cancer Res 68(2):589–596
125. Stebbins CE, Russo AA, Schneider C, Rosen N, Hartl FU, Pavletich NP (1997) Crystal
structure of an Hsp90-geldanamycin complex: targeting of a protein chaperone by an
antitumor agent. Cell 89(2):239–250
126. Park HJ, Rayalam S, Della-Fera MA, Ambati S, Yang JY, Baile CA (2008) Withaferin A
induces apoptosis and inhibits adipogenesis in 3T3-L1 adipocytes. Biofactors 33(2):137–148
127. Subramanian C, Zhang H, Gallagher R, Hammer G, Timmermann B, Cohen M (2014)
Withanolides are potent novel targeted therapeutic agents against adrenocortical carcinomas.
World J Surg 38(6):1343–1352
128. Samadi AK, Bazzill J, Zhang X, Gallagher R, Zhang H, Gollapudi R et al (2012) Novel
withanolides target medullary thyroid cancer through inhibition of both RET phosphorylation
and the mammalian target of rapamycin pathway. Surgery 152(6):1238–1247
14 Natural Withanolides in the Treatment of Chronic Diseases 367
129. Yadav VR, Prasad S, Sung B, Kannappan R, Aggarwal BB (2010) Targeting inflammatory
pathways by triterpenoids for prevention and treatment of cancer. Toxins 2:2428–2466
130. Zhang X, Mukerji R, Samadi AK, Cohen MS (2011) Down-regulation of estrogen
receptor-alpha and Rearranged during Transfection tyrosine kinase is associated with
Withaferin A-induced apoptosis in MCF-7 breast cancer cells. BMC Complement Altern
Med 11:84
131. Zhang X, Samadi AK, Roby KF, Timmermann B, Cohen MS (2012) Inhibition of cell
growth and induction of apoptosis in ovarian carcinoma cell lines CaOV3 and SKOV3 by
natural withanolide Withaferin A. Gynecol Oncol 124(3):606–612
132. Zhang X, Timmermann B, Samadi AK, Cohen MS (2012) Withaferin a induces
proteasome-dependent degradation of breast cancer susceptibility gene 1 and heat shock
factor 1 proteins in breast cancer cells. ISRN Biochem 2012:707586
133. Samadi AK, Cohen SM, Mukerji R, Chaguturu V, Zhang X, Timmermann BN et al (2012)
Natural withanolide withaferin A induces apoptosis in uveal melanoma cells by suppression
of Akt and c-MET activation. Tumor Biol 33(4):1179–1189
134. Cohen SM, Mukerji R, Timmermann BN, Samadi AK, Cohen MS (2012) A novel
combination of withaferin A and sorafenib shows synergistic efficacy against both papillary
and anaplastic thyroid cancers. Am J Surg 204(6):895–900; (discussion 900–891)
135. Motiwala HF, Bazzill J, Samadi A, Zhang H, Timmermann BN, Cohen MS et al (2013)
Synthesis and cytotoxicity of semisynthetic withalongolide A analogues. ACS Med Chem
Lett 4(11):1069–1073
136. Grogan PT, Sarkaria JN, Timmermann BN, Cohen MS (2014) Oxidative cytotoxic agent
withaferin A resensitizes temozolomide-resistant glioblastomas via MGMT depletion and
induces apoptosis through Akt/mTOR pathway inhibitory modulation. Invest New Drugs 32
(4):604–617
137. Vyas AR, Singh SV (2014) Molecular targets and mechanisms of cancer prevention and
treatment by withaferin A, a naturally occurring steroidal lactone. AAPS J 16(1):1–10
138. Grover A, Shandilya A, Agrawal V, Pratik P, Bhasme D, Bisaria VS et al (2011)
Hsp90/Cdc37 chaperone/co-chaperone complex, a novel junction anticancer target elucidated
by the mode of action of herbal drug withaferin A. BMC Bioinform 12(Suppl 1):S30
139. Gambhir L, Checker R, Sharma D, Thoh M, Patil A, Degani M et al (2015) Thiol dependent
NF-kappaB suppression and inhibition of T-cell mediated adaptive immune responses by a
naturally occurring steroidal lactone withaferin A. Toxicol Appl Pharmacol 289(2):297–312
140. Saha S, Islam MK, Shilpi JA, Hasan S (2013) Inhibition of VEGF: a novel mechanism to
control angiogenesis by Withania somnifera’s key metabolite withaferin A. In Silico
Pharmacol 1:11
141. White PT, Subramanian C, Zhu Q, Zhang H, Zhao H, Gallagher R et al (2016) Novel HSP90
inhibitors effectively target functions of thyroid cancer stem cell preventing migration and
invasion. Surgery 159(1):142–151
142. Lee DH, Lim I-H, Sung E-G, Kim J-Y, Song I-H, Park YK et al (2013) Withaferin A inhibits
matrix metalloproteinase-9 activity by suppressing the Akt signaling pathway. Oncol Rep 30
(2):933–938
143. Lee J, Hahm ER, Marcus AI, Singh SV (2015) Withaferin A inhibits experimental
epithelial-mesenchymal transition in MCF-10A cells and suppresses vimentin protein level
in vivo in breast tumors. Mol Carcinog 54(6):417–429
144. Lee J, Sehrawat A, Singh SV (2012) Withaferin A causes activation of Notch2 and Notch4 in
human breast cancer cells. Breast Cancer Res Treat 136(1):45–56
145. Amin H, Nayak D, Ur Rasool R, Chakraborty S, Kumar A, Yousuf K et al (2016) Par-4
dependent modulation of cellular beta-catenin by medicinal plant natural product derivative
3-azido withaferin A. Mol Carcinog 55(5):864–881
146. O’Connell MA, Hayes JD (2015) The Keap1/Nrf2 pathway in health and disease: from the
bench to the clinic. Biochem Soc Trans 43(4):687–689
368 P.T. White et al.
147. Hayes JD, Chowdhry S, Dinkova-Kostova AT, Sutherland C (2015) Dual regulation of
transcription factor Nrf2 by Keap1 and by the combined actions of beta-TrCP and GSK-3.
Biochem Soc Trans 43(4):611–620
148. Vaishnavi K, Saxena N, Shah N, Singh R, Manjunath K, Uthayakumar M et al (2012)
Differential activities of the two closely related withanolides, Withaferin A and Withanone:
bioinformatics and experimental evidences. PLoS ONE 7(9):e44419
149. Kostecka A, Sznarkowska A, Meller K, Acedo P, Shi Y, Mohammad Sakil HA et al (2014)
JNK-NQO1 axis drives TAp73-mediated tumor suppression upon oxidative and proteasomal
stress. Cell Death Dis 5:e1484
150. Scholz CC, Taylor CT (2013) Targeting the HIF pathway in inflammation and immunity.
Curr Opin Pharmacol 13(4):646–653
151. Arya M, Ahmed H, Silhi N, Williamson M, Patel HR (2007) Clinical importance and
therapeutic implications of the pivotal CXCL12-CXCR4 (chemokine ligand-receptor)
interaction in cancer cell migration. Tumour Biol 28(3):123–131
152. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100(1):57–70
153. Conway EM, Collen D, Carmeliet P (2001) Molecular mechanisms of blood vessel growth.
Cardiovasc Res 49(3):507–521
154. Laderoute KR, Calaoagan JM, Gustafson-Brown C, Knapp AM, Li GC, Mendonca HL et al
(2002) The response of c-jun/AP-1 to chronic hypoxia is hypoxia-inducible factor 1 alpha
dependent. Mol Cell Biol 22(8):2515–2523
155. Masoud GN, Li W (2015) HIF-1 pathway: role, regulation and intervention for cancer
therapy. Acta Pharm Sin B. 5(5):378–389
156. D’Ignazio L, Bandarra D, Rocha S (2016) NF-kappaB and HIF crosstalk in immune
responses. FEBS J 283(3):413–424
157. Semenza GL (2010) Defining the role of hypoxia-inducible factor 1 in cancer biology and
therapeutics. Oncogene 29(5):625–634
158. Vaupel P (2009) Prognostic potential of the pre-therapeutic tumor oxygenation status. Adv
Exp Med Biol 645:241–246
159. Gao R, Shah N, Lee JS, Katiyar SP, Li L, Oh E et al (2014) Withanone-rich combination of
Ashwagandha withanolides restricts metastasis and angiogenesis through hnRNP-K. Mol
Cancer Ther 13(12):2930–2940
160. Hitchon CA, El-Gabalawy HS (2004) Oxidation in rheumatoid arthritis. Arthritis Res Ther 6
(6):265–278
161. van’t Hof RJ, Hocking L, Wright PK, Ralston SH (2000) Nitric oxide is a mediator of
apoptosis in the rheumatoid joint. Rheumatology (Oxford) 39(9):1004–1008
162. Sumantran VN, Chandwaskar R, Joshi AK, Boddul S, Patwardhan B, Chopra A et al (2008)
The relationship between chondroprotective and antiinflammatory effects of Withania
somnifera root and glucosamine sulphate on human osteoarthritic cartilage in vitro. Phytother
Res 22(10):1342–1348
163. Subbaraju GV, Vanisree M, Rao CV, Sivaramakrishna C, Sridhar P, Jayaprakasam B et al
(2006) Ashwagandhanolide, a bioactive dimeric thiowithanolide isolated from the roots of
Withania somnifera. J Nat Prod 69(12):1790–1792
164. Wruck CJ, Fragoulis A, Gurzynski A, Brandenburg LO, Kan YW, Chan K et al (2011) Role
of oxidative stress in rheumatoid arthritis: insights from the Nrf2-knockout mice. Ann Rheum
Dis 70(5):844–850
165. Yu SM, Kim SJ (2013) Production of reactive oxygen species by withaferin A causes loss of
type collagen expression and COX-2 expression through the PI3K/Akt, p38, and JNK
pathways in rabbit articular chondrocytes. Exp Cell Res 319(18):2822–2834
166. Kim JH, Kim SJ (2014) Overexpression of microRNA-25 by withaferin A induces
cyclooxygenase-2 expression in rabbit articular chondrocytes. J Pharmacol Sci 125(1):83–90
167. Ganesan K, Sehgal PK, Mandal AB, Sayeed S (2011) Protective effect of Withania somnifera
and Cardiospermum halicacabum extracts against collagenolytic degradation of collagen.
Appl Biochem Biotechnol 165(3–4):1075–1091
14 Natural Withanolides in the Treatment of Chronic Diseases 369
168. Sumantran VN, Kulkarni A, Boddul S, Chinchwade T, Koppikar SJ, Harsulkar A et al (2007)
Chondroprotective potential of root extracts of Withania somnifera in osteoarthritis. J Biosci
32(2):299–307
169. Rasool M, Varalakshmi P (2007) Protective effect of Withania somnifera root powder in
relation to lipid peroxidation, antioxidant status, glycoproteins and bone collagen on
adjuvant-induced arthritis in rats. Fundam Clin Pharmacol 21(2):157–164
170. Khan MA, Subramaneyaan M, Arora VK, Banerjee BD, Ahmed RS (2015) Effect of
Withania somnifera (Ashwagandha) root extract on amelioration of oxidative stress and
autoantibodies production in collagen-induced arthritic rats. J Complement Integr Med 12
(2):117–125
171. Gupta A, Singh S (2014) Evaluation of anti-inflammatory effect of Withania somnifera root
on collagen-induced arthritis in rats. Pharm Biol 52(3):308–320
172. Rasool M, Varalakshmi P (2006) Immunomodulatory role of Withania somnifera root
powder on experimental induced inflammation: an in vivo and in vitro study. Vascul
Pharmacol 44(6):406–410
173. Rasool M, Varalakshmi P (2006) Suppressive effect of Withania somnifera root powder on
experimental gouty arthritis: an in vivo and in vitro study. Chem Biol Interact 164(3):174–180
174. Chopra A, Lavin P, Patwardhan B, Chitre D (2004) A 32-week randomized,
placebo-controlled clinical evaluation of RA-11, an Ayurvedic drug, on osteoarthritis of
the knees. J Clin Rheumatol 10(5):236–245
175. Kumar G, Srivastava A, Sharma SK, Rao TD, Gupta YK (2015) Efficacy & safety evaluation
of Ayurvedic treatment (Ashwagandha powder & Sidh Makardhwaj) in rheumatoid arthritis
patients: a pilot prospective study. Indian J Med Res 141(1):100–106
176. Gottschalk TA, Tsantikos E, Hibbs ML (2015) Pathogenic inflammation and its therapeutic
targeting in systemic lupus erythematosus. Front Immunol 6:550
177. Khor B, Gardet A, Xavier RJ (2011) Genetics and pathogenesis of inflammatory bowel
disease. Nature 474(7351):307–317
178. Yildirim-Toruner C, Diamond B (2011) Current and novel therapeutics in the treatment of
systemic lupus erythematosus. J Allergy Clin Immunol 127(2):303–312; (quiz 313–304)
179. Triantafillidis JK, Merikas E, Georgopoulos F (2011) Current and emerging drugs for the
treatment of inflammatory bowel disease. Drug Des Dev Ther 5:185–210
180. Minhas U, Minz R, Bhatnagar A (2011) Prophylactic effect of Withania somnifera on
inflammation in a non-autoimmune prone murine model of lupus. Drug Discov Ther 5
(4):195–201
181. Minhas U, Minz R, Das P, Bhatnagar A (2012) Therapeutic effect of Withania somnifera on
pristane-induced model of SLE. Inflammopharmacology 20(4):195–205
182. Samadi AK (2015) Potential anticancer properties and mechanisms of action of withanolides.
Enzymes 37:73–94
183. Dar NJ, Hamid A, Ahmad M (2015) Pharmacologic overview of Withania somnifera, the
Indian Ginseng. Cell Mol Life Sci 72(23):4445–4460
184. Widodo N, Priyandoko D, Shah N, Wadhwa R, Kaul SC (2010) Selective killing of cancer
cells by Ashwagandha leaf extract and its component withanone involves ROS signaling.
PLoS ONE 5(10):e13536
185. Nishikawa Y, Okuzaki D, Fukushima K, Mukai S, Ohno S, Ozaki Y et al (2015)
Withaferin A induces cell death selectively in androgen-independent prostate cancer cells but
not in normal fibroblast cells. PLoS ONE 10(7):e0134137
186. Park JW, Min KJ, Kim DE, Kwon TK (2015) Withaferin A induces apoptosis
through the generation of thiol oxidation in human head and neck cancer cells. Int J
Mol Med 35(1):247–252
187. Samadi AK, Mukerji R, Shah A, Timmermann BN, Cohen MS (2010) A novel RET inhibitor
with potent efficacy against medullary thyroid cancer in vivo. Surgery 148(6):1228–1236
188. Henrich CJ, Brooks AD, Erickson KL, Thomas CL, Bokesch HR, Tewary P et al (2015)
Withanolide E sensitizes renal carcinoma cells to TRAIL-induced apoptosis by increasing
cFLIP degradation. Cell Death Dis 6:e1666
370 P.T. White et al.
189. Thaiparambil JT, Bender L, Ganesh T, Kline E, Patel P, Liu Y et al (2011) Withaferin A
inhibits breast cancer invasion and metastasis at sub-cytotoxic doses by inducing vimentin
disassembly and serine 56 phosphorylation. Int J Cancer 129(11):2744–2755
190. Yang Z, Garcia A, Xu S, Powell DR, Vertino PM, Singh S et al (2013) Withania somnifera
root extract inhibits mammary cancer metastasis and epithelial to mesenchymal transition.
PLoS ONE 8(9):e75069
191. Su B-N, Gu J-Q, Kang Y-H, Park E-J, Pezzuto JM, Kinghorn AD (2004) Induction of the
phase II enzyme, quinone reductase, by withanolides and norwithanolides from solanaceous
species. Mini Rev Org Chem 1(1):115–123
192. Mathur S, Kaur P, Sharma M, Katyal A, Singh B, Tiwari M et al (2004) The treatment of skin
carcinoma, induced by UV B radiation, using 1-oxo-5beta, 6beta-epoxy-witha-2-enolide,
isolated from the roots of Withania somnifera, in a rat model. Phytomedicine 11(5):452–460
193. Padmavathi B, Rath PC, Rao AR, Singh RP (2005) Roots of Withania somnifera inhibit
forestomach and skin carcinogenesis in mice. Evid Based Complement Altern Med 2
(1):99–105
194. Li W, Zhang C, Du H, Huang V, Sun B, Harris JP et al (October 2015) Withaferin A
suppresses the up-regulation of acetyl-coA carboxylase 1 and skin tumor formation in a skin
carcinogenesis mouse model. Mol Carcinog 1–8. doi: 10.1002/mc.22423. (Epub ahead of
print)
195. Hahm ER, Lee J, Kim SH, Sehrawat A, Arlotti JA, Shiva SS et al (2013) Metabolic
alterations in mammary cancer prevention by withaferin A in a clinically relevant mouse
model. J Natl Cancer Inst 105(15):1111–1122
196. Kim SH, Singh SV (2014) Mammary cancer chemoprevention by withaferin A is
accompanied by in vivo suppression of self-renewal of cancer stem cells. Cancer Prev Res
(Phila) 7(7):738–747
197. Khazal KF, Hill DL, Grubbs CJ (2014) Effect of Withania somnifera root extract on
spontaneous estrogen receptor-negative mammary cancer in MMTV/Neu mice. Anticancer
Res 34(11):6327–6332
198. Kataria H, Kumar S, Chaudhary H, Kaur G (2016) Withania somnifera suppresses tumor
growth of intracranial allograft of glioma cells. Mol Neurobiol 53(6):4143–4158
199. McKenna MK, Gachuki BW, Alhakeem SS, Oben KN, Rangnekar VM, Gupta RC et al
(2015) Anti-cancer activity of withaferin A in B-cell lymphoma. Cancer Biol Ther 16
(7):1088–1098
200. Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK et al (2014)
Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of
ovarian cancer by targeting putative cancer stem cells. PLoS ONE 9(9):e107596
201. Fong MY, Jin S, Rane M, Singh RK, Gupta R, Kakar SS (2012) Withaferin A synergizes the
therapeutic effect of doxorubicin through ROS-mediated autophagy in ovarian cancer.
PLoS ONE 7(7):e42265
202. Yang H, Shi G, Dou QP (2007) The tumor proteasome is a primary target for the natural
anticancer compound Withaferin A isolated from “Indian winter cherry”. Mol Pharmacol 71
(2):426–437
203. Srinivasan S, Ranga RS, Burikhanov R, Han SS, Chendil D (2007) Par-4-dependent
apoptosis by the dietary compound withaferin A in prostate cancer cells. Cancer Res 67
(1):246–253
204. Lahat G, Zhu QS, Huang KL, Wang S, Bolshakov S, Liu J et al (2010) Vimentin is a novel
anti-cancer therapeutic target; insights from in vitro and in vivo mice xenograft studies.
PLoS ONE 5(4):e10105
205. Munagala R, Kausar H, Munjal C, Gupta RC (2011) Withaferin A induces p53-dependent
apoptosis by repression of HPV oncogenes and upregulation of tumor suppressor proteins in
human cervical cancer cells. Carcinogenesis 32(11):1697–1705
206. Yang H, Wang Y, Cheryan VT, Wu W, Cui CQ, Polin LA et al (2012) Withaferin A inhibits
the proteasome activity in mesothelioma in vitro and in vivo. PLoS ONE 7(8):e41214
14 Natural Withanolides in the Treatment of Chronic Diseases 371
207. Biswal BM, Sulaiman SA, Ismail HC, Zakaria H, Musa KI (2013) Effect of Withania
somnifera (Ashwagandha) on the development of chemotherapy-induced fatigue and quality
of life in breast cancer patients. Integr Cancer Ther 12(4):312–322
208. Amor S, Peferoen LA, Vogel DY, Breur M, van der Valk P, Baker D et al (2014)
Inflammation in neurodegenerative diseases–an update. Immunology 142(2):151–166
209. Amor S, Woodroofe MN (2014) Innate and adaptive immune responses in neurodegeneration
and repair. Immunology 141(3):287–291
210. Zhao J, Nakamura N, Hattori M, Kuboyama T, Tohda C, Komatsu K (2002) Withanolide
derivatives from the roots of Withania somnifera and their neurite outgrowth activities. Chem
Pharm Bull (Tokyo) 50(6):760–765
211. Kuboyama T, Tohda C, Komatsu K (2005) Neuritic regeneration and synaptic reconstruction
induced by withanolide A. Br J Pharmacol 144(7):961–971
212. Martorana F, Guidotti G, Brambilla L, Rossi D (2015) Withaferin A inhibits nuclear
factor-kappaB-dependent pro-inflammatory and stress response pathways in the astrocytes.
Neural Plast 2015:381964
213. Patil SP, Maki S, Khedkar SA, Rigby AC, Chan C (2010) Withanolide A and asiatic acid
modulate multiple targets associated with amyloid-beta precursor protein processing and
amyloid-beta protein clearance. J Nat Prod 73(7):1196–1202
214. Choudhary MI, Yousuf S, Nawaz SA, Ahmed S, Atta ur R (2004) Cholinesterase inhibiting
withanolides from Withania somnifera. Chem Pharm Bull (Tokyo) 52(11):1358–1361
215. Riaz N, Malik A, Nawaz SA, Muhammad P, Choudhary MI (2004) Cholinesterase-inhibiting
withanolides from Ajuga bracteosa. Chem Biodivers 1(9):1289–1295
216. Grover A, Shandilya A, Agrawal V, Bisaria VS, Sundar D (2012) Computational evidence to
inhibition of human acetyl cholinesterase by withanolide a for Alzheimer treatment. J Biomol
Struct Dyn 29(4):651–662
217. Durg S, Dhadde SB, Vandal R, Shivakumar BS, Charan CS (2015) Withania somnifera
(Ashwagandha) in neurobehavioural disorders induced by brain oxidative stress in rodents: a
systematic review and meta-analysis. J Pharm Pharmacol 67(7):879–899
218. Sehgal N, Gupta A, Valli RK, Joshi SD, Mills JT, Hamel E et al (2012) Withania somnifera
reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein
receptor-related protein in liver. Proc Natl Acad Sci 109(9):3510–3515
219. Galvan A, Wichmann T (2008) Pathophysiology of parkinsonism. Clin Neurophysiol 119
(7):1459–1474
220. Przedborski S, Levivier M, Jiang H, Ferreira M, Jackson-Lewis V, Donaldson D et al (1995)
Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal
injection of 6-hydroxydopamine. Neuroscience 67(3):631–647
221. Przedborski S, Vila M (2003) The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse
model: a tool to explore the pathogenesis of Parkinson’s disease. Ann N Y Acad Sci
991:189–198
222. Prakash J, Yadav SK, Chouhan S, Singh SP (2013) Neuroprotective role of Withania
somnifera root extract in maneb-paraquat induced mouse model of parkinsonism. Neurochem
Res 38(5):972–980
223. RajaSankar S, Manivasagam T, Sankar V, Prakash S, Muthusamy R, Krishnamurti A et al
(2009) Withania somnifera root extract improves catecholamines and physiological
abnormalities seen in a Parkinson’s disease model mouse. J Ethnopharmacol 125(3):369–373
224. Ahmad M, Saleem S, Ahmad AS, Ansari MA, Yousuf S, Hoda MN et al (2005)
Neuroprotective effects of Withania somnifera on 6-hydroxydopamine induced Parkinsonism
in rats. Hum Exp Toxicol 24(3):137–147
225. RajaSankar S, Manivasagam T, Surendran S (2009) Ashwagandha leaf extract: a potential
agent in treating oxidative damage and physiological abnormalities seen in a mouse model of
Parkinson’s disease. Neurosci Lett 454(1):11–15
226. Sankar SR, Manivasagam T, Krishnamurti A, Ramanathan M (2007) The neuroprotective
effect of Withania somnifera root extract in MPTP-intoxicated mice: an analysis of
behavioral and biochemical variables. Cell Mol Biol Lett 12(4):473–481
372 P.T. White et al.
227. Prakash J, Chouhan S, Yadav SK, Westfall S, Rai SN, Singh SP (2014) Withania somnifera
alleviates parkinsonian phenotypes by inhibiting apoptotic pathways in dopaminergic
neurons. Neurochem Res 39(12):2527–2536
228. Crotti A, Glass CK (2015) The choreography of neuroinflammation in Huntington’s disease.
Trends Immunol 36(6):364–373
229. Kumar P, Kumar A (2008) Effects of root extract of Withania somnifera in
3-nitropropionic acid-induced cognitive dysfunction and oxidative damage in rats. Int J
Health Res 1(3):139–149
230. Kumar P, Kumar A (2009) Possible neuroprotective effect of Withania somnifera root extract
against 3-nitropropionic acid-induced behavioral, biochemical, and mitochondrial
dysfunction in an animal model of Huntington’s disease. J Med Food 12(3):591–600
231. Nagashayana N, Sankarankutty P, Nampoothiri MR, Mohan PK, Mohanakumar KP (2000)
Association of L-DOPA with recovery following Ayurveda medication in Parkinson’s
disease. J Neurol Sci 176(2):124–127
232. Tian R, Hou G, Li D, Yuan TF (2014) A possible change process of inflammatory cytokines
in the prolonged chronic stress and its ultimate implications for health. Sci World J
2014:780616
233. Furtado M, Katzman MA (2015) Neuroinflammatory pathways in anxiety, posttraumatic
stress, and obsessive compulsive disorders. Psychiatry Res 229(1–2):37–48
234. Raison CL, Capuron L, Miller AH (2006) Cytokines sing the blues: inflammation and the
pathogenesis of depression. Trends Immunol 27(1):24–31
235. Tohmi M, Tsuda N, Watanabe Y, Kakita A, Nawa H (2004) Perinatal inflammatory cytokine
challenge results in distinct neurobehavioral alterations in rats: implication in psychiatric
disorders of developmental origin. Neurosci Res 50(1):67–75
236. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E (2008) Inflammatory cytokine
alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry 63(8):801–808
237. Goldstein BI, Kemp DE, Soczynska JK, McIntyre RS (2009) Inflammation and the
phenomenology, pathophysiology, comorbidity, and treatment of bipolar disorder: a
systematic review of the literature. J Clin Psychiatry 70(8):1078–1090
238. Berk M, Kapczinski F, Andreazza AC, Dean OM, Giorlando F, Maes M et al (2011)
Pathways underlying neuroprogression in bipolar disorder: focus on inflammation, oxidative
stress and neurotrophic factors. Neurosci Biobehav Rev 35(3):804–817
239. Gray SM, Bloch MH (2012) Systematic review of proinflammatory cytokines in obsessive–
compulsive disorder. Curr Psychiatry Rep 14(3):220–228
240. Rege NN, Thatte UM, Dahanukar SA (1999) Adaptogenic properties of six rasayana herbs
used in Ayurvedic medicine. Phytother Res 13(4):275–291
241. Panossian A, Wikman G (2009) Evidence-based efficacy of adaptogens in fatigue, and
molecular mechanisms related to their stress-protective activity. Curr Clin Pharmacol 4
(3):198–219
242. Prud’homme GJ, Glinka Y, Wang Q (2015) Immunological GABAergic interactions and
therapeutic applications in autoimmune diseases. Autoimmun Rev 14(11):1048–1056
243. Mehta AK, Binkley P, Gandhi SS, Ticku MK (1991) Pharmacological effects of Withania
somnifera root extract on GABAA receptor complex. Indian J Med Res 94:312–315
244. Candelario M, Cuellar E, Reyes-Ruiz JM, Darabedian N, Feimeng Z, Miledi R et al (2015)
Direct evidence for GABAergic activity of Withania somnifera on mammalian ionotropic
GABAA and GABArho receptors. J Ethnopharmacol 171:264–272
245. Bhattacharya SK, Muruganandam AV (2003) Adaptogenic activity of Withania somnifera:
an experimental study using a rat model of chronic stress. Pharmacol Biochem Behav 75
(3):547–555
246. Gupta GL, Rana AC (2007) Protective effect of Withania somnifera dunal root extract against
protracted social isolation induced behavior in rats. Indian J Physiol Pharmacol 51(4):345–353
247. Gupta GL, Rana AC (2008) Effect of Withania somnifera Dunal in ethanol-induced
anxiolysis and withdrawal anxiety in rats. Indian J Exp Biol 46(6):470–475
14 Natural Withanolides in the Treatment of Chronic Diseases 373
Abstract Kokum, a spice derived from the fruit of the Garcinia hanburyi tree, is
traditionally used in Ayurvedic medicines to facilitate digestion and to treat sores,
dermatitis, diarrhoea, dysentery, and ear infection. One of the major active com-
ponents of kokum is gambogic acid, also known as guttic acid, guttatic acid,
beta-guttilactone, and beta-guttiferin. Gambogic acid’s anti-proliferative,
anti-bacterial; antioxidant and anti-inflammatory effects result from its modulation
of numerous cell-signaling intermediates. This chapter discusses the sources,
chemical components, mechanism of action, and disease targets of the kokum spice.
Keywords Neutraceuticals Dietary agents Gambogic acid Kokum Cancer
Signal transduction pathways
15.1 Introduction
Mother Nature has gifted us a variety of natural agents, including nutraceuticals. One
of the well-known nutraceuticals is Gambogic acid (GA), which is a xanthonoid
derived from the brownish or orange resin from Garcinia hanburyi (Fig. 15.1).
Garcinia hanburyi is a small to medium-sized evergreen tree with smooth gray bark,
and it is native to Cambodia, southern Vietnam, and Thailand. Garcinia indica,
primarily of Indian origin, is known by many names: bindin, biran, bhirand, bhinda,
kokum, katambi, panarpuli, ratamba, amsol, and tamal. In English language, it is
commonly known as mangosteen, wild mangosteen, red mango, Hanbury’s Garcinia,
gambojia, gamboge, and Indian gamboge tree. Germans called this gummi-gutti.
The Garcinia indica seed contains 23–26 % oil, which is used in confectionery,
medicines, and cosmetics. It is used in curries and other dishes as a slightly bitter
spice, a souring agent, and as a substitute for tamarind.
Gambogic Acid
Fig. 15.1 Plant species and fruits by which gambogic acid is derived. Highlighted circles on GA
structure indicate the most common sites for novel derivative generation
Enthusiasm shown by researchers from around the globe clearly suggests that GA
has been one of the “hot” nutraceuticals. GA has shown to be effective on different
chronic diseases (Sect. 4.1 covers different chronic diseases), but its effect on
cancer has been studied the most. Almost a decade ago our group showed that the
anti-inflammatory and anticancer response of GA is associated with its inhibitory
response on Nuclear Factor-Kappa B (NF-jB) [7], since then plethora of studies
suggest that GA regulates several key signaling pathways. GA exhibits
anti-proliferative, antioxidant, and anti-inflammatory effects by modulating cell
signaling pathways, enzymes, and molecular targets, such as epigenetic regulators,
protein kinases, transcription factors, inflammatory biomarkers, and growth regu-
lators. Through microarray analysis, GA modulates many gene products [8, 9]
(Table 15.1).
The transcription factor NF-jB is one of the major mediators of inflammation and is
linked with many diseases including cancer, diabetes, arthritis, and neurological
disorders. Therefore, an agent that can suppress NF-jB activation has potential for
clinical use against various chronic illnesses. GA suppression of NF-jB activation
induced by TNF-a, LPS, and various agents [7, 10] leads to the suppression of
NF-jB regulated products, such as cyclooxygenase type 2 (COX-2), inducible
nitric oxide synthase (iNOS), and survival proteins [7, 10, 11]. These actions give it
great potential as a broad-spectrum clinical agent.
MAPKs are evolutionarily conserved enzymes that play a key role in the inflam-
matory stimuli and environmental stresses that lead to the activation of three
independent pathways: p44/42 MAPK extracellular signal-regulated kinases 1 and
2 (ERK1/ERK2), c-Jun N-terminal kinase, and p38 MAPK [20]. In vitro studies of
several cancer cells showed that GA inhibits MAPK pathways [21]. Moreover, this
phytochemical also inhibited ERK in HT-29, HepG2, KBM5, and NCI-H460
cancer cells [22–24].
The Src family of proteins consists of eight non-receptor tyrosine kinases charac-
terized by a common structure [25]. Src kinases are involved in signal transduction
pathways that are triggered by a variety of surface receptors, including receptors for
tyrosine kinases, integrin, and antigens, as well as receptors coupled with the
G-protein [25, 26]. As a consequence of changes observed in protein expression
and kinase activity in cancer cells, the Src family has been implicated in the
development of cancer [25, 26]. This prompted the design of specific inhibitors, the
most common of which are adenine mimetics, to treat solid tumors and leukemia
clinically [25, 26]. In addition, some of the Src kinases expressed in hematopoietic
cells play pivotal roles in lymphocyte maturation and activation [25]. This finding
encouraged the development of safe and effective Src-specific inhibitors that are
currently in clinical trials as immune-suppressants for the treatment of immuno-
logical disorders [15, 27]. Separate research showing that GA inhibits Src in PC3,
and K562 cells suggests that GA may also have clinical potential against cancers
and immunological disorders in which Src plays a pivotal role [15, 16].
Proteins in the STAT family are among the best studied of the latent cytoplasmic
signal-dependent transcription factors [28–30]. In vitro studies of the MCF-7,
MCF-10A, U266, and MM1.s cell lines suggest that GA modulates the nuclear
translocation and DNA binding of STAT3 and inhibits genes modulated by this
transcription factor [28, 31].
380 M.K. Pandey et al.
ROS have been linked with various cell signaling pathways [54, 55]. GA, induces
the production of ROS [56].
Overexpression of COX-2 is associated with many cancers and is linked with tumor
cell proliferation and suppression of apoptosis [57, 58]. Therefore, COX-2 inhibi-
tors have great potential in the treatment of cancers and inflammatory conditions, as
evidenced by the U. S. Food and Drug Administration’s approval of celecoxib, a
known COX-2 inhibitor, for the treatment of various inflammatory conditions [59].
GA, too, has been shown to inhibit COX-2 activation induced by TNF-a in KBM5
leukemic cells [7].
382 M.K. Pandey et al.
PARPs are cell signaling enzymes present in eukaryotes and are involved in poly
(ADP ribosylation) of DNA-binding proteins [73]. Pharmacological degradation of
PARP-1 may enhance the activity of antitumor drugs by inhibiting necrosis and
15 Gambogic Acid and Its Role in Chronic Diseases 383
activating apoptosis [74]. In vitro studies have shown that GA induces PARP
degradation and enhances apoptosis in T98 glioma, HeLa, non-small lung cancer
A549 and NCI-H460, breast cancer, and multiple myeloma cells [7, 11, 17, 22, 24,
31, 66, 75–77].
The bcl-2 gene family consists of at least 25 genes that are proapoptotic or
anti-apoptotic and share at least one of the four characteristic BH domains [81]. The
anti-apoptotic protein bcl-2, which displays sequence homology in all four domains
(i.e., BH1–BH4), promotes cell survival [82]. Increased expression of the bcl-2
protein commonly occurs in human malignancies and is associated with disease
maintenance and progression, resistance to chemotherapy, and poor clinical out-
come. Antisense oligonucleotides targeting bcl-2 have been shown to facilitate
apoptosis in various tumor types [83]. Therefore, bcl-2 inhibitors have great
potential in the treatment of cancer. In vitro and in vivo studies showed that GA
inhibits bcl-2 expression in MGC-803, HL-60, MCF-7, A375M, SMMC-7721,
BGC-823, Jeko-1, and K562 [15, 84–88].
BAD is proapoptotic and proliferative, suggesting that the cell cycle functions of the
multi-domain bcl-2 family members [89]. BAD antagonizes both the cell cycle and
anti-apoptotic functions of bcl-2 and bcl-xL through BH3 binding [89].
Overexpression of the BH3-only molecule BAD renders the cell unable to arrest in G0
and persistently activates cdk2 [92]. Previous study showed that GA in combination
with nanoparticle Fe3O4 activates BAD and induces apoptosis in LOVO cells [93].
Caspases play a central role in mediating various apoptotic responses. In vitro and
in vivo research of GA has shown that it induces the activation of caspase-3 and
caspase-9 in various cancer cells including glioma, osteosarcoma, non-small lung
cancer, leukemia, lymphoma, breast cancer, pancreatic cancer, melanoma and
multiple myeloma and, induces apoptosis [7, 11, 17, 22, 24, 31, 66, 75–77].
Extensive studies from past one decade have shed light on GA’s potential as
anti-inflammatory and anticancer agents. So far the focus of the studies have been to
identify the molecular targets by which GA exerts its effects, primarily on cancer
cells. However, a very recent study showed that GA could be used as an
anti-psoriatic agent [97]. Importantly, the molecular mechanism by which GA
mediates its effect strongly suggest that it could be used for the prevention and
treatment of many organ and tissue disorders, which are associated with inflam-
mation and oxidative stress. GA alleviates oxidative stress, inflammation in chronic
diseases and regulates inflammatory and pro-inflammatory pathways related with
most chronic diseases.
15 Gambogic Acid and Its Role in Chronic Diseases 385
Type 2 diabetes is a chronic disease where cells have reduced insulin signaling,
leading to hyperglycemia, and long-term complications, such as heart, kidney, and
liver disease. Recently, more and more studies have shown the critical roles of
oxidative stress and inflammatory reactions in the pathogenesis of diabetes. Studies
have shown that AMP-activated protein kinase (AMPK) plays a key role in
maintaining intracellular and whole-body energy homeostasis. Activation of AMPK
has been shown to ameliorate the symptoms of type 2 diabetes and obesity. In vitro
studies by Zhao et al. [107] demonstrate that GA, activates AMPK by increasing the
phosphorylation of AMPKa and its downstream substrate ACC in various cell lines
[107]. This group also showed that GA induced activation of AMPK was associated
with increased intracellular ROS level. Collectively, these results suggest that GA
may be a novel direct activator of AMPK and could be used as anti-diabetic agent.
However, further studies are required to fully evaluate this function of GA.
15.4.4 Psoriasis
15.4.5 Cancer
Inflammation plays key roles in all the ways of tumorigenesis and therapy response
[110]. Activation and interaction between STAT3 and NF-jB are very vital in the
control of cancer cells and inflammatory cells [111]. TNF-a, VEGF, IL-10, MMP-2
and MMP-9, MCP, CD4+ T, AP-1, Akt, PPAR-c, MAP kinases, and mTORC1 are
also important linking factors between inflammation and cancer [111]. It has been
shown that GA suppresses the growth of various cancer cells such as non-small cell
lung cancer [112], human hepatocellular carcinoma [113], oral squamous cell
carcinoma [114], human breast cancer [86], human malignant melanoma [115],
human gastric carcinoma [116], and human leukemia cancer [7] and multiple
myeloma [31, 117]. A variety of mechanisms have been proposed by which GA
inhibits the proliferation of cancer cells and induces apoptosis. These include
inhibition of antiapoptotic proteins Bcl-2 [86, 88] and survivin [118]; induction of
apoptosis-associated proteins p53 [119], bax, and procaspase-3 [115]; activation of
15 Gambogic Acid and Its Role in Chronic Diseases 387
MM cells
MCL-1
BCL-2
NF-κB BCL-xL
Gambogic IAPs
acid Cyclin D Survival
IL-6 Anti-apoptosis
MCL-1 Proliferation
JAK/STAT3 Inflammation
BCL-xL
Migration
Raf MEK/ERK Drug resistance
IL-6, VEGF,
TNF-α, PI3K
SDF-1α NF-κB
etc. mTOR
Akt Capspase9
VEGF, TGF
TFs Cytokines
Growth factors
BMSCs
Fig. 15.2 Intimate relationship between multiple myeloma and bone marrow microenvironment.
Bone marrow stromal cells secrete cytokine a growth factors, these growth factors activates several
pathways in multiple myeloma. Gambogic acid targets these pathways
388 M.K. Pandey et al.
Table 15.2 A list of studies describing antitumor effects of gambogic acid in animals
Tumor Cell line Route Dose (mg/kg) Model References
Lung SPC-A1 i.v. 4, 8 Xenograft [121]
NCI-H1993 i.p. 10, 20 or 30 Xenograft [127]
NCI-H1975 i.v. 8 Xenograft [137]
A549 i.p. 8, 16 and 32 Xenograft [134]
Gastric cancer BGC-823 i.v. 8 Xenograft [85]
Cervical carcinoma HeLa cells i.p. 2 Xenograft [128]
Chronic myeloid KBM5 i.p. 3 Xenograft [24]
leukemia
Liver HepG2 i.v. 1.5 Xenograft [132]
Ovarian SKOV3 1.0 Xenograft [133]
Breast MDA-MB-231 i.v. 4 and 8 Orthotopic [94]
Hepatoma H22 i.v. 2, 4 and 8 Xenograft [125]
H22 p.o. 12.5, 25, 30 and Xenograft [125]
50
SMMC-7721 i.v. 2, 4, and 8 Xenograft [131, 136]
Melanoma B16-F10 i.v. 0.375, 0.75, and Orthotopic [135]
1.5
Colon HT-29 i.v. 5, 10 and 20 Xenograft [129]
15 Gambogic Acid and Its Role in Chronic Diseases 389
15.7 Conclusions
The spice derived from kokum, the fruit of Garcinia indica, is used in Indian
cuisines and Ayurvedic medicine. The main component isolated from kokum is
GA, which demonstrates thrust quencher, antioxidant, antimicrobial, antiulceration,
and anticancer properties. Although GA is a potent, biologically active compound,
only a number of studies are carried out in animals and none have been done in
humans. Because of its diverse range of biological activity in vitro, more in vivo
and clinical studies are warranted to establish its true usefulness as a clinical
therapeutic agent in a variety of human diseases.
References
1. Guo Q et al (2006) Toxicological studies of gambogic acid and its potential targets in
experimental animals. Basic Clin Pharmacol Toxicol 99(2):178–184
2. Noguer O, Villena J, Lorita J, Vilaro S, Reina M (2009) Syndecan-2 downregulation impairs
angiogenesis in human microvascular endothelial cells. Exp Cell Res 315(5):795–808
3. Jang SW et al (2007) Gambogic amide, a selective agonist for TrkA receptor that possesses
robust neurotrophic activity, prevents neuronal cell death. Proc Natl Acad Sci USA 104
(41):16329–16334
4. Qi Q et al (2008) Studies on the toxicity of gambogic acid in rats. J Ethnopharmacol 117
(3):433–438
5. Udvadia AJ, Linney E (2003) Windows into development: historic, current, and future
perspectives on transgenic zebrafish. Dev Biol 256(1):1–17
6. Zhang HZ et al (2004) Discovery, characterization and SAR of gambogic acid as a potent
apoptosis inducer by a HTS assay. Bioorg Med Chem 12(2):309–317
7. Pandey MK et al (2007) Gambogic acid, a novel ligand for transferrin receptor, potentiates
TNF-induced apoptosis through modulation of the nuclear factor-kappaB signaling pathway.
Blood 110(10):3517–3525
8. Li X et al (2013) Gambogic acid is a tissue-specific proteasome inhibitor in vitro and in vivo.
Cell Rep 3(1):211–222
9. Wang Y et al (2014) Methyl jasmonate sensitizes human bladder cancer cells to gambogic
acid-induced apoptosis through down-regulation of EZH2 expression by miR-101. Br J
Pharmacol 171(3):618–635
10. Palempalli UD et al (2009) Gambogic acid covalently modifies IkappaB kinase-beta subunit
to mediate suppression of lipopolysaccharide-induced activation of NF-kappaB in
macrophages. Biochem J 419(2):401–409
390 M.K. Pandey et al.
11. Yang LJ, Chen Y (2013) New targets for the antitumor activity of gambogic acid in
hematologic malignancies. Acta Pharmacol Sin 34(2):191–198
12. Franke TF (2008) PI3K/Akt: getting it right matters. Oncogene 27(50):6473–6488
13. Franke TF, Hornik CP, Segev L, Shostak GA, Sugimoto C (2003) PI3K/Akt and apoptosis:
size matters. Oncogene 22(56):8983–8998
14. Fruman DA, Rommel C (2014) PI3K and cancer: lessons, challenges and opportunities. Nat
Rev Drug Discov 13(2):140–156
15. Li R et al (2009) Gambogic acid induces G0/G1 arrest and apoptosis involving inhibition of
SRC-3 and inactivation of Akt pathway in K562 leukemia cells. Toxicology 262(2):98–105
16. Yi T et al (2008) Gambogic acid inhibits angiogenesis and prostate tumor growth by
suppressing vascular endothelial growth factor receptor 2 signaling. Cancer Res 68(6):1843–
1850
17. Pandey MK et al (2014) Gambogic acid inhibits multiple myeloma mediated
osteoclastogenesis through suppression of chemokine receptor CXCR4 signaling
pathways. Exp Hematol 42(10):883–896
18. Yang Y, Sun X, Yang Y, Yang X, Zhu H, Dai S, Chen X, Zhang H, Guo Q, Song Y,
Wang F, Cheng H, Sun X (2016) Gambogic acid enhances the radiosensitivity of human
esophageal cancer cells by inducing reactive oxygen species via targeting Akt/mTOR
pathway. Tumour Biol 37(2):1853–1862
19. Ma J et al (2015) Gambogic acid inhibits osteoclast formation and ovariectomy-induced
osteoporosis by suppressing the JNK, p38 and Akt signalling pathways. Biochem J 469
(3):399–408
20. Chen J et al (2008) Microtubule depolymerization and phosphorylation of c-Jun N-terminal
kinase-1 and p38 were involved in gambogic acid induced cell cycle arrest and apoptosis in
human breast carcinoma MCF-7 cells. Life Sci 83(3–4):103–109
21. Lu N et al (2007) Gambogic acid inhibits angiogenesis through suppressing vascular
endothelial growth factor-induced tyrosine phosphorylation of KDR/Flk-1. Cancer Lett 258
(1):80–89
22. Wang LH et al (2014) Gambogic acid synergistically potentiates cisplatin-induced apoptosis
in non-small-cell lung cancer through suppressing NF-kappaB and MAPK/HO-1 signalling.
Br J Cancer 110(2):341–352
23. Yan F et al (2012) Gambogenic acid induced mitochondrial-dependent apoptosis and referred
to phospho-Erk1/2 and phospho-p38 MAPK in human hepatoma HepG2 cells. Environ
Toxicol Pharmacol 33(2):181–190
24. Shi X et al (2014) Gambogic acid induces apoptosis in imatinib-resistant chronic myeloid
leukemia cells via inducing proteasome inhibition and caspase-dependent Bcr-Abl
downregulation. Clin Cancer Res 20(1):151–163
25. Parsons SJ, Parsons JT (2004) Src family kinases, key regulators of signal transduction.
Oncogene 23(48):7906–7909
26. Benati D, Baldari CT (2008) SRC family kinases as potential therapeutic targets for
malignancies and immunological disorders. Curr Med Chem 15(12):1154–1165
27. Aleshin A, Finn RS (2010) SRC: a century of science brought to the clinic. Neoplasia 12
(8):599–607
28. Yu H, Jove R (2004) The STATs of cancer—new molecular targets come of age. Nat Rev
Cancer 4(2):97–105
29. Turkson J, Jove R (2000) STAT proteins: novel molecular targets for cancer drug discovery.
Oncogene 19(56):6613–6626
30. Clevenger CV (2004) Roles and regulation of stat family transcription factors in human
breast cancer. Am J Pathol 165(5):1449–1460
31. Prasad S, Pandey MK, Yadav VR, Aggarwal BB (2011) Gambogic acid inhibits STAT3
phosphorylation through activation of protein tyrosine phosphatase SHP-1: potential role in
proliferation and apoptosis. Cancer Prev Res 4(7):1084–1094
32. Pandey MK, Rastogi S, Kale VP, Gowda T, Amin SG (2014) Targeting CXCL12/CXCR4
axis in multiple myeloma. J Hematol Thrombo Dis 2:159
15 Gambogic Acid and Its Role in Chronic Diseases 391
58. Greenhough A et al (2009) The COX-2/PGE2 pathway: key roles in the hallmarks of cancer
and adaptation to the tumour microenvironment. Carcinogenesis 30(3):377–386
59. Tindall E (1999) Celecoxib for the treatment of pain and inflammation: the preclinical and
clinical results. J Am Osteopath Assoc 99(11 Suppl):S13–S17
60. Page-McCaw A, Ewald AJ, Werb Z (2007) Matrix metalloproteinases and the regulation of
tissue remodelling. Nat Rev Mol Cell Biol 8(3):221–233
61. Parks WC, Wilson CL, Lopez-Boado YS (2004) Matrix metalloproteinases as modulators of
inflammation and innate immunity. Nat Rev Immunol 4(8):617–629
62. Egeblad M, Werb Z (2002) New functions for the matrix metalloproteinases in cancer
progression. Nat Rev Cancer 2(3):161–174
63. Gialeli C, Theocharis AD, Karamanos NK (2011) Roles of matrix metalloproteinases in
cancer progression and their pharmacological targeting. FEBS J 278(1):16–27
64. Shay G, Lynch CC, Fingleton B (2015) Moving targets: emerging roles for MMPs in cancer
progression and metastasis. Matrix Biol 44–46:200–206
65. Qi Q et al (2008) Involvement of matrix metalloproteinase 2 and 9 in gambogic acid induced
suppression of MDA-MB-435 human breast carcinoma cell lung metastasis. J Mol Med 86
(12):1367–1377
66. Qi Q et al (2008) Anti-invasive effect of gambogic acid in MDA-MB-231 human breast
carcinoma cells. Biochem Cell Biol 86(5):386–395
67. Etienne-Manneville S (2010) From signaling pathways to microtubule dynamics: the key
players. Curr Opin Cell Biol 22(1):104–111
68. Dumontet C, Jordan MA (2010) Microtubule-binding agents: a dynamic field of cancer
therapeutics. Nat Rev Drug Discov 9(10):790–803
69. Jordan MA, Wilson L (2004) Microtubules as a target for anticancer drugs. Nat Rev Cancer 4
(4):253–265
70. Hochegger H, Takeda S, Hunt T (2008) Cyclin-dependent kinases and cell-cycle transitions:
does one fit all? Nat Rev Mol Cell Biol 9(11):910–916
71. Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL (2011) Cyclin D as a
therapeutic target in cancer. Nat Rev Cancer 11(8):558–572
72. Hosokawa Y, Arnold A (1998) Mechanism of cyclin D1 (CCND1, PRAD1) overexpression
in human cancer cells: analysis of allele-specific expression. Genes Chromosom Cancer 22
(1):66–71
73. Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG (2010) PARP inhibition:
PARP1 and beyond. Nat Rev Cancer 10(4):293–301
74. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA (2008) DNA repair pathways
as targets for cancer therapy. Nat Rev Cancer 8(3):193–204
75. Krajarng A et al (2015) Apoptosis induction associated with the ER stress response through
up-regulation of JNK in HeLa cells by gambogic acid. BMC Complement Altern Med 15:26
76. Thida M, Kim DW, Tran TT, Pham MQ, Lee H, Kim I, Lee JW (2016) Gambogic acid
induces apoptotic cell death in T98G glioma cells. Bioorg Med Chem Lett 26(3):1097–1101
77. Yang LJ et al (2012) Effects of gambogic acid on the activation of caspase-3 and
downregulation of SIRT1 in RPMI-8226 multiple myeloma cells via the accumulation of
ROS. Oncol Lett 3(5):1159–1165
78. Wang X, Lin Y (2008) Tumor necrosis factor and cancer, buddies or foes? Acta Pharmacol
Sin 29(11):1275–1288
79. Lee JY, Lee BH, Lee JY (2015) Gambogic acid disrupts toll-like receptor4 activation by
blocking lipopolysaccharides binding to myeloid differentiation factor 2. Toxicol Res 31
(1):11–16
80. Liao CH, Sang S, Liang YC, Ho CT, Lin JK (2004) Suppression of inducible nitric oxide
synthase and cyclooxygenase-2 in downregulating nuclear factor-kappa B pathway by
Garcinol. Mol Carcinog 41(3):140–149
81. Youle RJ, Strasser A (2008) The BCL-2 protein family: opposing activities that mediate cell
death. Nat Rev Mol Cell Biol 9(1):47–59
15 Gambogic Acid and Its Role in Chronic Diseases 393
82. Juin P, Geneste O, Gautier F, Depil S, Campone M (2013) Decoding and unlocking the
BCL-2 dependency of cancer cells. Nat Rev Cancer 13(7):455–465
83. Gleave ME, Monia BP (2005) Antisense therapy for cancer. Nat Rev Cancer 5(6):468–479
84. Xu J et al (2013) Gambogic acid induces mitochondria-dependent apoptosis by modulation
of Bcl-2 and Bax in mantle cell lymphoma JeKo-1 cells. Chin J Cancer Res 25(2):183–191
85. Liu W et al (2005) Anticancer effect and apoptosis induction of gambogic acid in human
gastric cancer line BGC-823. World J Gastroenterol 11(24):3655–3659
86. Gu H et al (2009) Gambogic acid reduced bcl-2 expression via p53 in human breast MCF-7
cancer cells. J Cancer Res Clin Oncol 135(12):1777–1782
87. Zhai D et al (2008) Gambogic acid is an antagonist of antiapoptotic Bcl-2 family proteins.
Mol Cancer Ther 7(6):1639–1646
88. Zhao L, Guo QL, You QD, Wu ZQ, Gu HY (2004) Gambogic acid induces apoptosis and
regulates expressions of Bax and Bcl-2 protein in human gastric carcinoma MGC-803 cells.
Biol Pharm Bull 27(7):998–1003
89. Czabotar PE, Lessene G, Strasser A, Adams JM (2014) Control of apoptosis by the BCL-2
protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol 15(1):49–63
90. Ma SB et al (2014) Bax targets mitochondria by distinct mechanisms before or during
apoptotic cell death: a requirement for VDAC2 or Bak for efficient Bax apoptotic function.
Cell Death Differ 21(12):1925–1935
91. Xie H et al (2009) GA3, a new gambogic acid derivative, exhibits potent antitumor activities
in vitro via apoptosis-involved mechanisms. Acta Pharmacol Sin 30(3):346–354
92. Zinkel S, Gross A, Yang E (2006) BCL2 family in DNA damage and cell cycle control. Cell
Death Differ 13(8):1351–1359
93. Fang L et al (2012) Synergistic effect of a combination of nanoparticulate Fe3O4 and
gambogic acid on phosphatidylinositol 3-kinase/Akt/Bad pathway of LOVO cells. Int J
Nanomed 7:4109–4118
94. Li C et al (2012) Gambogic acid promotes apoptosis and resistance to metastatic potential in
MDA-MB-231 human breast carcinoma cells. Biochem Cell Biol 90(6):718–730
95. Ishaq M et al (2014) Gambogic acid induced oxidative stress dependent caspase activation
regulates both apoptosis and autophagy by targeting various key molecules (NF-kappaB,
Beclin-1, p62 and NBR1) in human bladder cancer cells. Biochim Biophys Acta 1840
(12):3374–3384
96. Tang C et al (2009) Downregulation of survivin and activation of caspase-3 through the
PI3K/Akt pathway in ursolic acid-induced HepG2 cell apoptosis. Anticancer Drugs 20
(4):249–258
97. Wen J et al (2014) Gambogic acid exhibits anti-psoriatic efficacy through inhibition of
angiogenesis and inflammation. J Dermatol Sci 74(3):242–250
98. Costa S, Reina-Couto M, Albino-Teixeira A, Sousa T (2016) Statins and oxidative stress in
chronic heart failure. Rev Port J Cardiol 35(1):41–57
99. Urbieta Caceres VH et al (2011) Early experimental hypertension preserves the myocardial
microvasculature but aggravates cardiac injury distal to chronic coronary artery obstruction.
Am J Physiol Heart Circ Physiol 300(2):H693–H701
100. Liu S et al (2013) Gambogic acid suppresses pressure overload cardiac hypertrophy in rats.
Am J Cardiovasc Dis 3(4):227–238
101. McInnes IB, Schett G (2011) The pathogenesis of rheumatoid arthritis. N Engl J Med 365
(23):2205–2219
102. Wruck CJ et al (2011) Role of oxidative stress in rheumatoid arthritis: insights from the
Nrf2-knockout mice. Ann Rheum Dis 70(5):844–850
103. Ray PD, Huang BW, Tsuji Y (2012) Reactive oxygen species (ROS) homeostasis and redox
regulation in cellular signaling. Cell Signal 24(5):981–990
104. Kahlenberg JM, Fox DA (2011) Advances in the medical treatment of rheumatoid arthritis.
Hand Clin 27(1):11–20
105. Forestier R et al (2009) Non-drug treatment (excluding surgery) in rheumatoid arthritis:
clinical practice guidelines. Joint Bone Spine 76(6):691–698
394 M.K. Pandey et al.
106. Cascao R et al (2014) Potent anti-inflammatory and antiproliferative effects of gambogic acid
in a rat model of antigen-induced arthritis. Mediat Inflamm 2014:195327
107. Zhao B, Shen H, Zhang L, Shen Y (2012) Gambogic acid activates AMP-activated protein
kinase in mammalian cells. Biochem Biophys Res Commun 424(1):100–104
108. Gupta MA, Simpson FC, Gupta AK (2015) Psoriasis and sleep disorders: a systematic
review. Sleep Med Rev 29:63–75
109. Coimbra S, Figueiredo A, Castro E, Rocha-Pereira P, Santos-Silva A (2012) The roles of
cells and cytokines in the pathogenesis of psoriasis. Int J Dermatol 51(4):389–395; quiz 395–
388
110. Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G (2006) Inflammation and
cancer: how hot is the link? Biochem Pharmacol 72(11):1605–1621
111. Grivennikov SI, Karin M (2010) Dangerous liaisons: STAT3 and NF-kappaB collaboration
and crosstalk in cancer. Cytokine Growth Factor Rev 21(1):11–19
112. Zhu X et al (2009) Mechanisms of gambogic acid-induced apoptosis in non-small cell lung
cancer cells in relation to transferrin receptors. J Chemother 21(6):666–672
113. Mu R et al (2010) An oxidative analogue of gambogic acid-induced apoptosis of human
hepatocellular carcinoma cell line HepG2 is involved in its anticancer activity in vitro. Eur J
Cancer Prev 19(1):61–67
114. He D et al (2009) The NF-kappa B inhibitor, celastrol, could enhance the anti-cancer effect of
gambogic acid on oral squamous cell carcinoma. BMC Cancer 9:343
115. Xu X et al (2009) Gambogic acid induces apoptosis by regulating the expression of Bax and
Bcl-2 and enhancing caspase-3 activity in human malignant melanoma A375 cells. Int J
Dermatol 48(2):186–192
116. Wang X et al (2009) Proteomic identification of molecular targets of gambogic acid: role of
stathmin in hepatocellular carcinoma. Proteomics 9(2):242–253
117. Wang F et al (2014) Gambogic acid suppresses hypoxia-induced hypoxia-inducible
factor-1alpha/vascular endothelial growth factor expression via inhibiting
phosphatidylinositol 3-kinase/Akt/mammalian target protein of rapamycin pathway in
multiple myeloma cells. Cancer Sci 105(8):1063–1070
118. Wang T et al (2008) Gambogic acid, a potent inhibitor of survivin, reverses docetaxel
resistance in gastric cancer cells. Cancer Lett 262(2):214–222
119. Rong JJ et al (2010) Gambogic acid triggers DNA damage signaling that induces p53/p21
(Waf1/CIP1) activation through the ATR-Chk1 pathway. Cancer Lett 296(1):55–64
120. Qin Y et al (2007) Gambogic acid inhibits the catalytic activity of human topoisomerase
IIalpha by binding to its ATPase domain. Mol Cancer Ther 6(9):2429–2440
121. Wu ZQ, Guo QL, You QD, Zhao L, Gu HY (2004) Gambogic acid inhibits proliferation of
human lung carcinoma SPC-A1 cells in vivo and in vitro and represses telomerase activity
and telomerase reverse transcriptase mRNA expression in the cells. Biol Pharm Bull 27
(11):1769–1774
122. Yu J et al (2006) Repression of telomerase reverse transcriptase mRNA and hTERT promoter
by gambogic acid in human gastric carcinoma cells. Cancer Chemother Pharmacol 58
(4):434–443
123. Kasibhatla S et al (2005) A role for transferrin receptor in triggering apoptosis when targeted
with gambogic acid. Proc Natl Acad Sci USA 102(34):12095–12100
124. Pandey MK, Kale VP, Song C, Sung SS, Sharma AK, Talamo G, Dovat S, Amin SG (2014)
Gambogic acid inhibits multiple myeloma mediated osteoclastogenesis through suppression
of chemokine receptor CXCR4 signaling pathways. Exp Hematol 42(10):883–896
125. Gu H et al (2008) Gambogic acid induced tumor cell apoptosis by T lymphocyte activation in
H22 transplanted mice. Int Immunopharmacol 8(11):1493–1502
126. Chi Y et al (2013) An open-labeled, randomized, multicenter phase IIa study of gambogic
acid injection for advanced malignant tumors. Chin Med J 126(9):1642–1646
127. Li D et al (2015) Antitumor activity of gambogic acid on NCI-H1993 xenografts via MET
signaling pathway downregulation. Oncol Lett 10(5):2802–2806
15 Gambogic Acid and Its Role in Chronic Diseases 395
128. Yue Q et al (2016) proteomic analysis revealed the important role of vimentin in human
cervical carcinoma HeLa cells treated with gambogic acid. MCP 15(1):26–44
129. Huang GM, Sun Y, Ge X, Wan X, Li CB (2015) Gambogic acid induces apoptosis and
inhibits colorectal tumor growth via mitochondrial pathways. WJG 21(20):6194–6205
130. Shi X et al (2015) Gambogic acid induces apoptosis in diffuse large B-cell lymphoma cells
via inducing proteasome inhibition. Sci Rep 5:9694
131. Yang Y et al (2007) Differential apoptotic induction of gambogic acid, a novel anticancer
natural product, on hepatoma cells and normal hepatocytes. Cancer Lett 256(2):259–266
132. Lu N et al (2013) Gambogic acid inhibits angiogenesis through inhibiting
PHD2-VHL-HIF-1alpha pathway. Eur J Pharm Sci 49(2):220–226
133. Wang J, Yuan Z (2013) Gambogic acid sensitizes ovarian cancer cells to doxorubicin
through ROS-mediated apoptosis. Cell Biochem Biophys 67(1):199–206
134. Li Q et al (2010) Gambogenic acid inhibits proliferation of A549 cells through
apoptosis-inducing and cell cycle arresting. Biol Pharm Bull 33(3):415–420
135. Zhao J et al (2008) Inhibition of alpha(4) integrin mediated adhesion was involved in the
reduction of B16-F10 melanoma cells lung colonization in C57BL/6 mice treated with
gambogic acid. Eur J Pharmacol 589(1–3):127–131
136. Guo QL, You QD, Wu ZQ, Yuan ST, Zhao L (2004) General gambogic acids inhibited
growth of human hepatoma SMMC-7721 cells in vitro and in nude mice. Acta Pharmacol Sin
25(6):769–774
137. Wang C, Wang W, Wang C, Tang Y, Tian H (2015) Combined therapy with EGFR TKI and
gambogic acid for overcoming resistance in -T790M mutant lung cancer. Oncol Lett 10
(4):2063–2066
Chapter 16
Embelin and Its Role in Chronic Diseases
Hong Lu, Jun Wang, Youxue Wang, Liang Qiao and Yongning Zhou
Abstract Embelia ribes Burm of Myrsinaceae family has been widely used as an
herb in the traditional medicine of India. Embelin is an active component extracted
from the fruits of Embelia ribes. It has a wide spectrum of biological activities and
is not toxic at low dose. This review focuses on the physical–chemical properties
and bioactivities of Embelin, as well as its effects on chronic diseases such as
tumors, autoimmune inflammatory diseases, parasitic infections, microbial infec-
tions, diabetes, obesity, and cardio-cerebral vascular diseases. The underlying
mechanisms of the effects are also discussed. As a multiple-targeted therapeutic
agent, Embelin has the potential to be used widely for the treatment of a variety of
chronic diseases, including malignant tumors.
Keywords Embelin Anticancer activity Autoimmune inflammatory diseases
Anthelmintic activity Antimicrobial activity Diabetes Cardio-cerebral vascular
diseases
16.1 Introduction
H OH
HO CH 2 - C 9 H 18 - CH 3
Fig. 16.1 Constitutional formula of Embelin (Molecular formula C17H26O4; Molecular weight
294.39; Melting point 142–143 °C; Appearance Orange solid, Log p (octanol-water) 4.34;
Solubility Insoluble in water, but soluble in organic solvents such as DMSO and Ether) [89]
16 Embelin and Its Role in Chronic Diseases 399
Embelin is a phenolic lipid that can be found as a secondary metabolite not only in
plants, but also in fungi, bacteria, and animals both during normal development and
in response to stress conditions such as infection, wounding, and UV radiation
[115]. It is structurally similar to natural coenzyme Q10 (ubiquinones), and it has
antioxidant activity similar to that of coenzyme Q10.
Embelin is also a weak mitochondrial uncoupler that dissociates the electron
transport system from ADP phosphorylation and inhibits ATP synthesis [79]. The
uncoupling effect also leads to increased oxygen consumption. It may explain why
Embelin caused swelling of the mitochondria in rat liver and respiratory inhibition
in germinated cowpea. It can also explain why Embelin prevented angiogenesis
during tumor growth and wound healing by exhausting the low respiratory reserve
of proliferating endothelial cells through uncoupling action [22].
Although Embelin possesses several promising biological activities, preclinical
efforts have been hampered because of water insolubility which leads to poor
bioavailability by oral route. In order to resolve the solubility issue and improve
biological activity, new techniques, such as utilization of the liquisolid compact
systems [95], formulation of Embelin–phospholipid complex (EPC) [98], con-
struction of Embelin-loaded thermosensitive injectable hydrogel system [99, 100],
and synthesis of Embelin analogues [18, 28, 69, 128] have been tried. Polymeric
micelle technique has also been introduced to enhancing the water solubility and
bioavailability [27].
400 H. Lu et al.
Some progresses have been made to improve the solubility and delivery of
Embelin. A micelle system was developed through the conjugation of Embelin and
polyethylene glycol (PEG) through an aspartic acid bridge. The resulting
PEG-Embelin micelles is dual functional and can efficiently deliver both Embelin
itself and other hydrophobic antitumor drugs, such as Paclitaxel, which are
encapsulated in the micelles. This system has been tested with success in cancer cell
line in vitro and animal cancer models in vivo [51, 76]. Diammonium salt of
Embelin exists in some plants and can easily be made from purified Embelin. It was
found to be highly soluble in water and its biological activities were found to be
similar to those of Embelin [28]. It could be an ideal therapeutic agent which can be
administered easily.
Toxicity studies revealed that Embelin is safe to use. It did not show toxic effect
on human fibroblasts at 20 µg/ml for 72 h in vitro [104]. In the acute toxicity study,
no prominent signs of toxicity or death were observed in mice administered orally
with crude hydroalcoholic extract of Embelia ribes at the dose up to 5000 mg/kg
for 24 h [28]. In chronic toxicity study, the administration of Embelin at the dose of
50 mg/kg/day for 14 weeks to Wistar rats did not cause any drastic drop in the
blood counts [132]. In another study, administration of Embelin at 10 mg/kg/day
for 10 weeks in rats did not show toxic side effects on bone marrow, liver, kidney,
and heart [111]. Embelin at doses of 50 mg and 100 mg/kg of body weight/day for
14 days did not cause significant body weight changes, mortality, or apparent signs
of toxic effects in mice [115]. Previous studies had also reported the nontoxic nature
of Embelin on hematopoietic cells when administered for 6 months in mice, rats,
and monkeys [55].
The specifics and details of some biomedicinal activities of Embelin will also be
discussed in the following sections.
Anticancer effect of Embelin has been studied in various kinds of cancer, including
breast cancer [29, 133], prostate cancer [94, 106, 107], hepatic cancer [131, 132],
pancreatic cancer [86, 99, 100], colon cancer [24, 25], gastric cancer [141], leu-
kemia [49, 50], and multiple myeloma [47], through in vivo and in vitro experi-
ments. It can effectively inhibit tumor cell proliferation and migration, induce tumor
cell apoptosis, and inhibit tumor invasion, metastasis and angiogenesis.
The molecular mechanisms of the anticancer effect of Embelin have also been
studied extensively. Multiple signaling pathways have been found to be involved.
Embelin was predicted to be a strong inhibitor of XIAP through structure-based
computational screening of a traditional herbal medicine three-dimensional struc-
ture database [92]. It also downregulates the expression of XIAP [27, 50, 86, 141].
16 Embelin and Its Role in Chronic Diseases 401
XIAP belongs to inhibitor of apoptosis (IAP) family [119]. It binds to and inhibits
the initiator caspase-9 and effector caspase-3, caspase-7, and consequently prevents
intrinsic apoptotic pathways [114, 126]. XIAP is overexpressed in various types of
cancer cells [136]. Embelin could induce apoptosis of tumor cell through down-
regulating expression of XIAP and activating caspase cascades [27, 50, 141]. It
could also transform TRAIL-resistant pancreatic cancer cells into TRAIL-sensitive
cells by suppressing the expression of XIAP in vitro [86].
Nuclear factor-jB (NF-jB) plays an important role in tumor cell survival and
proliferation, as well as tumor angiogenesis, invasion, and inflammation [3, 8, 101].
Embelin blocks NF-jB signaling pathway resulting in the suppression of NF-jB
regulated antiapoptotic and metastatic gene products. Embelin inhibits the
expression of ICAM-1, MMP-9, COX-2, cyclin D1, c-Myc, and VEGF [25, 122,
141], all of which are regulated by NF-jB and closely related to proliferation,
invasion, and angiogenesis of tumors. It also inhibits NF-jB-dependent apoptosis
gene products including survivin, XIAP, IAP-1/2, TRAF1, cFLIP and Bcl-2, and
downregulates Bcl-xL [65, 94].
Researches showed Embelin suppressed activation of NF-jB not through
modifying NF-jB proteins itself but through inhibiting IKK (IjB kinase) activa-
tion, IjBa (the inhibitor of NF-jB)-phosphorylation and degradation, and p65
phosphorylation and acetylation, which then inhibited nuclear translocation of
NF-jB and expression of the NF-jB-dependent reporter genes [3, 112, 145]. IKK
could phosphorylate and degrade IjBa [38]. When IjBa was degraded, NF-jB
translocated to the nucleus and activated the transcription of specific genes [41].
These results suggested that Embelin induces apoptosis not only by inhibiting and
downregulating XIAP, but also through suppressing NF-jB signaling. XIAP, as a
downstream target gene, could be activated by NF-jB [63].
Embelin also regulates activity of the PI3 K/AKT pathway by reducing AKT
expression levels, and increases PTEN levels in cancer cell lines in vitro. The
PI3 K/Akt pathway contributes to tumor formation through the anti-apoptotic
activity of Akt. Akt inhibits apoptosis through phosphorylation of GSK3b, which
leads to inactivation of itself [56] and maintains the stabilization of b-catenin [71].
Embelin could suppress b-catenin expression through inhibition of activation of
AKT and GSK-3b, and thus promote apoptosis [94]. Suppression of Akt activation
could also lead to P53 activation and turn on pro-apoptotic signaling pathways for
induction of apoptosis [42]. At the same time, AKT could induce activation of
transcriptional factors, such as NF-jB [72]. Further research indicated that AKT
could activate IKK, and then cause NF-jB activation and cell survival [61]. The
upregulated PTEN can suppress activity of AKT [17].
Embelin upregulates the expression of P53 both in vivo and in vitro [99, 100,
133, 141]. P53 protein, as a key regulator of apoptosis, controls cell cycle pro-
gression and DNA repair, and induces apoptosis by regulating the expression of
proteins like Bcl-2 and Bax, which concomitantly induce Cytochrome C release and
activate Caspase cascades (primarily Caspase 9 and Caspase 3) leading to apoptosis
[130].
402 H. Lu et al.
Fig. 16.2 Schematic diagrams showing antitumor mechanism of Embelin. EB Embelin, XIAP
X-chromosome linked inhibitor of apoptosis protein, NF-jB Nuclear factor-jB, IjBa the inhibitor
of NF-jB, IKK IjB kinase, AKT protein kinase B, GSK3b glycogen synthase kinase-3b, JAK
janus-like kinase, PPARc peroxisome proliferator-activated receptor gamma, mTOR mammalian
target of rapamycin, S6K1 ribosomal S6 kinase 1, STAT3 signal transducer and activator of
transcription 3
effective therapeutic agents with few side effects. Embelia ribes have been shown to
relieve the symptoms of fever and skin diseases for many years. The effects of
Embelin on ulcerative colitis, psoriasis, autoimmune encephalomyelitis, rheumatoid
arthritis and the underlying mechanisms have also been studied.
Embelin was found to be effective in treating acetic acid-induced ulcerative
colitis in rats. It relieved symptoms such as body weight loss, diarrhea, and gross
intestinal bleeding. It decreased wet weight of the colon tissue [7] and alleviated
colonic shortening and splenomegaly [68], indicating the improvement of the
inflammation of colon. It decreased the colonic myeloperoxidase (MPO) activity
and lipid peroxides (LPO), as well as serum lactate dehydrogenase (LDH). It also
16 Embelin and Its Role in Chronic Diseases 405
increased the reduced glutathione (GSH) in this rat ulcerative colitis model [7, 137,
138]. Wet weight of the colon tissue is considered as a reliable indicator of the
extent of the inflammation in colitis. MPO exists abundantly in the neutrophils, and
the levels of colonic MPO reflect the infiltration of activated neutrophils in colon
which in turn reflect the severity of the colitis [21]. LPO is the product of membrane
lipid peroxidation, and it can cause further lipid peroxidation and generate more
reactive metabolites through self propagation chain reaction. LPO exhausts cellular
antioxidants and accelerates the development of inflammation and ulceration. GSH
can block free radical damage, enhance the antioxidant activity, facilitate the
transport of amino acids, and play a critical role in detoxification [44]. The ele-
vation of LDH in serum indicates enhanced production of lactic acid [80] and
increased release of LDH by damaged cells.
Pro-inflammatory cytokines (TNF-a, IFN-c, IL-1b, IL-6 and IL-12), and
anti-inflammatory cytokines (IL-4, IL-10, IL-11) play a central role during the
occurrence and development of IBD [46, 121]. TNF-a can disrupt the epithelial
barrier, induce the apoptosis of epithelial cells, and stimulate the secretions of
chemokines from intestinal epithelial cells. It can also activate immune system of
the bowel through recruiting and activating neutrophils and macrophages [15].
IL-1b can increase the infiltration of inflammatory cells into the intestine. Embelin
was found to ameliorated colitis by suppressing the level of TNF-a, IL-1b, and IL-6
in the colonic tissues of mice with dextran sulfate sodium (DSS)-induced colitis
[68].
Epidermal keratinocytes are known to participate in immune and inflammatory
reactions by producing a variety of cytokines including TNF-a, which plays a
crucial role in autoimmune diseases of skin, such as psoriasis. TNF-a released from
keratinocytes promotes the accumulation of inflammatory cells. Embelin could
inhibit topical edema, and decrease skin thickness, tissue weight, and MPO activity
in phorbol ester (TPA)-induced inflammation in mouse ear, which serves as a
model of psoriasis. The possible mechanism might be the blockage of the pro-
duction of pro-inflammatory cytokines (TNF-a, IL-1b) by keratinocytes [60].
Experimental Autoimmune Encephalomyelitis (EAE) is an autoimmune disorder
of the CNS that serves as an animal model for human MS [66]. Dendritic cells
(DCs) play a definitive role in the induction and maintenance of self-tolerance, and
the absence of these cells can lead to autoimmune inflammatory diseases, such as
MS. Transforming growth factor-beta (TGF-b) signaling in DCs was found to be a
prerequisite for the control of autoimmune encephalomyelitis [146], and it could
program DCs into a tolerogenic state [16], and thereby limit the inflammatory
response and promote the recovery from EAE [52]. Embelin was shown to sig-
nificantly increase TGF-b/b-catenin signaling, decrease STAT3 phosphorylation in
DCs, maintain DCs in their tolerogenic state, and block the expression of
pro-inflammatory factors (IFN-c, IL-12, IL-6, and IL-23), which lead to the
improvement of the EAE clinical score and alleviation of CNS inflammation and
demyelination [144].
The effect of Embelin on rheumatoid arthritis was also affirmed. Embelin was
found to reduce inflammation and bone erosion in a mouse model of inflammatory
406 H. Lu et al.
Embelia ribes Burm has been used as an anthelminthic remedy in India for a long
time. The fruits of Embelia ribes are widely used to expel the adult stage beef
tapeworm and other intestinal parasites [23, 39]. The ethanolic extract from the
seeds of Embelia ribes (10–200 lg/mL) exhibited potent anthelmintic activity
against roundworm Rhabditis pseudoelongata in vitro [43]. Crude hydroalcoholic
extract from the fruits showed significant anthelmintic activities against the dwarf
tapeworm (Hymenolepis nana) in vivo and against hookworm larva and Necator
americanus in vitro [28]. Diammonium salt of Embelin showed anthelmintic
activity against intestinal cestodes (Hymenolepis diminuta and Hymenolepis
microstoma), trematode (Echinostoma caproni), nematode (Heligmosomoides
polygyrus), and hookworm larva in vitro. The in vivo anthelmintic activity of
diammonium salt of Embelin against Hymenolepis diminuta and Hymenolepis nana
was also confirmed. However, the anthelmintic activity against Hymenolepis
microstoma, Echinostoma caproni, or Heligmosomoides polygyrus was not con-
firmed [6, 28]. In addition, Embelin was moderately active against Leishmania
amazonensis and Leishmania braziliensis lysing 70 % of the parasites at
100 µg/ml, and potently active against Trypanosoma cruzi trypomastigotes with
100 % lysis at 100 µg/ml in vitro [34].
The crude extracts from Embelia ribes have displayed moderate antibacterial
activity against Salmonella typhi, Staphylococcus aureus, Enterobacter aerogenes,
and Klebsiella pneumoniae [5, 109, 135].
Embelin was found to suppress Escherichia coli and methicillin-sensitive and
methicillin-resistant strains of Staphylococcus aureus with minimal inhibitory
concentration(MIC) values of 50, 250, and 62 lg/ml respectively [34]. At high
concentration (100 lg/disk), it significantly inhibited the growth of Staphylococcus
aureus, Streptococcus pyogenes, Shigella flexneri, Shigella sonnei, and
Pseudomonas aeruginosa; and was moderately effective against Salmonella typhi,
Shigella boydii, and Proteus mirabilis [19]. Further study indicated Embelin
showed bactericidal activity against Gram positive organisms, and bacteriostatic
activity against Gram negative organisms [108].
16 Embelin and Its Role in Chronic Diseases 407
It has been proved that Embelin was effective in the treatment of seizure, epi-
lepsy, and anxiety in animals. It has anticonvulsant activity against both grand mal
and petit mal epilepsy. In animal experiment, Embelin inhibited seizures induced by
maximal electroshock and pentylenetetrazole in a dose-dependent manner. The
anticonvulsant activity of Embelin was comparable to that of phenytoin and dia-
zepam [77, 78]. Embelin also showed a dose-dependent anxiolytic effect in animal
experiment [1].
16.5 Conclusion
Acknowledgments This work was supported by the grant “The Model for Prevention and
Control of Malignant Tumor in Area with High Incidence from Wuwei Gansu Province” (National
Science and Technology Program, Grant Code: 2012GS620101 to YN Zhou).
References
6. B∅gh HO, Andreassen JR, Lemmich J (1996) Anthelmintic usage of extracts of Embelia
schimperi from Tanzania. J Ethnopharmacol 50:35–42
7. Badamaranahalli SS, Kopparam M, Bhagawati ST, Durg S (2015) Embelin lipid nanospheres
for enhanced treatment of ulcerative colitis–Preparation, characterization and in vivo
evaluation. Eur J Pharm Sci 76:73–82
8. Baker RG, Hayden MS, Ghosh S (2011) NF-jB, inflammation, and metabolic disease. Cell
Metab 13:11–22
9. Bartlett JMS (2010) Biomarkers and Patient Selection for PI3K/Akt/mTOR targeted
therapies: current status and future directions. Clin Breast Cancer 10:S86–S95
10. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK (2006)
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and
regulatory T cells. Nature 441:235–238
11. Bhandari U, Ansari MN, Islam F (2008) Cardioprotective effect of aqueous extract of
Embelia ribes Burm fruits against isoproterenol-induced myocardial infarction in albino rats.
Indian J Exp Biol 9:35–40
12. Bhandari U, Chaudhari HS, Bisnoi AN, Kumar V, Khanna G, Javed K (2013) Anti-obesity
effect of standardized ethanol extract of Embelia ribes in murine model of high fat
diet-induced obesity. PharmaNutrition 1:50–57
13. Bhandari U, Jain N, Ansari MN, Pillai KK (2008) Beneficial effect of Embelia ribes ethanolic
extract on blood pressure and glycosylated hemoglobin in streptozotocin-induced diabetes in
rats. Fitoterapia 79:351–355
14. Bhandari U, Jain N, Pillai KK (2007) Further studies on antioxidant potential and protection
of pancreatic beta-cells by Embelia ribes in experimental diabetes. Exp Diabetes Res 2007
(ID: 15803)
15. Bischoff SC, Lorentz A, Schwengberg S, Weier G, Raab R, Manns MP (1999) Mast cells are
an important cellular source of tumour necrosis factor alpha in human intestinal tissue. Gut
44:643–652
16. Bryan VL, Zachary TB, Robert CF, Nir H, James JC, Marianne B (2011) TGF-b suppresses
b-catenin-dependent tolerogenic activation program in dendritic cells. PLoS ONE 6:e20099
17. Cantley LC, Neel BG (1999) New insights into tumor suppression: PTEN suppresses tumor
formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci
96:4240–4245
18. Chen J, Nikolovska-Coleska Z, Wang G, Qiu S, Wang S (2006) Design, synthesis, and
characterization of new embelin derivatives as potent inhibitors of X-linked inhibitor of
apoptosis protein. Bioorg Med Chem Lett 16:5805–5808
19. Chitra M, Shyamala Devi CS, Sukumar E (2003) Antibacterial activity of embelin.
Fitoterapia 74(4):401–403
20. Chitra M, Sukumar E, Suja V, Devi CSS (1994) Antitumor, anti-Inflammatory and analgesic
property of embelin, a plant product. Chemotherapy 40:109–113
21. Choudhary S, Keshavarzian A, Yong S, Wade M, Bocckino S, Day BJ, Banan A (2001)
Novel antioxidants zolimid and AEOL11201 ameliorate colitis in rats. Dig Dis Sci 46:2222–
2230
22. Coutelle O, Hornig-Do HT, Witt A, Andree M, Schiffmann LM, Piekarek M, Brinkmann K,
Seeger JM, Liwschitz M, Miwa S, Hallek M, Kronke M, Trifunovic A, Eming SA,
Wiesner RJ, Hacker UT, Kashkar H (2014) Embelin inhibits endothelial mitochondrial
respiration and impairs neoangiogenesis during tumor growth and wound healing. EMBO
Mol Med 6:624–639
23. D’Avigdor E, Wohlmuth H, Asfaw Z, Awas T (2014) The current status of knowledge of
herbal medicine and medicinal plants in Fiche, Ethiopia. J Ethnobiol Ethnomed 10:1–33
24. Dai Y, Jiao H, Teng G, Wang W, Zhang R, Wang Y, Hebbard L, George J, Qiao L (2014)
Embelin reduces colitis-associated tumorigenesis through limiting IL-6/STAT3 signaling.
Mol Cancer Ther 13:1206–1216
412 H. Lu et al.
25. Dai Y, Qiao L, Chan KW, Yang M, Ye J, Ma J, Zou B, Gu Q, Wang J, Pang R, Lan HY,
Wong BC (2009) Peroxisome proliferator-activated receptor-gamma contributes to the
inhibitory effects of Embelin on colon carcinogenesis. Cancer Res 69:4776–4783
26. Danquah M, Duke CB 3rd, Patil R, Miller DD, Mahato RI (2012) Combination therapy of
antiandrogen and XIAP inhibitor for treating advanced prostate cancer. Pharm Res 29:2079–
2091
27. Danquah M, Li F, Duke CB 3rd, Miller DD, Mahato RI (2009) Micellar delivery of
bicalutamide and embelin for treating prostate cancer. Pharm Res 26:2081–2092
28. Debebe Y, Tefera M, Mekonnen W, Abebe D, Woldekidan S, Abebe A, Belete Y,
Menberu T, Belayneh B, Tesfaye B, Nasir I, Yirsaw K, Basha H, Dawit A, Debella A (2015)
Evaluation of anthelmintic potential of the Ethiopian medicinal plant Embelia schimperi
Vatke in vivo and in vitro against some intestinal parasites. BMC Complement Altern Med
15:187
29. Dhanjal JK, Nigam N, Sharma S, Chaudhary A, Kaul SC, Grover A, Wadhwa R (2014)
Embelin inhibits TNF-a converting enzyme and cancer cell metastasis: molecular dynamics
and experimental evidence. BMC Cancer 14:775
30. Dharmapatni AA, Cantley MD, Marino V, Perilli E, Crotti TN, Smith MD, Haynes DR
(2015) The X-linked inhibitor of apoptosis protein inhibitor embelin suppresses inflammation
and bone erosion in collagen antibody induced arthritis mice. Mediators Inflamm 2015(2):
1–10 (ID: 564042)
31. Dowling RJ, Sonenberg N (2010) Downstream of mTOR: translational control of cancer. In:
mTOR pathway and mTOR inhibitors in cancer therapy. Humana Press, pp. 201–216.
32. Dwivedi D, Singh V (2015) Effects of the natural compounds embelin and piperine on the
biofilm-producing property of Streptococcus mutans. J Tradit Complement Med. http://www.
elsevier.com/locate/jtcme
33. Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, Van Obberghen E (2000)
SOCS-3 is an insulin-induced negative regulator of insulin signaling. J Biol Chem
275:15985–15991
34. Feresin GE, Tapia A, Sortino M, Zacchino S, Arias ARd, Inchausti A, Yaluff G, Rodriguez J,
Theoduloz C, Schmeda-Hirschmann G (2003) Bioactive alkyl phenols and embelin from
Oxalis erythrorhiza. J Ethnopharmacol 88:241–247
35. Ferreria GM, Laddha KS (2013) Histochemical localization of embelin in fruits of Embellia
Ribes Brum and its quantification. Int J Pharma Biosci Technol 1:16–19
36. Gandhi GR, Stalin A, Balakrishna K, Ignacimuthu S, Paulraj MG, Vishal R (2013) Insulin
sensitization via partial agonism of PPARgamma and glucose uptake through translocation
and activation of GLUT4 in PI3K/p-Akt signaling pathway by embelin in type 2 diabetic rats.
Biochim Biophys Acta 1830:2243–2255
37. Ghazi H, Hirotsugu M, Norio N, Masao H, Nobuko K, Kunitada S (2000) Inhibitory effects
of sudanese medicinal plant extracts on hepatitis C virus (HCV) protease. Phytother Res
14:510–516
38. Ghosh S, Karin M (2002) Missing pieces in the NF-jB puzzle. Cell 109:S81–S96
39. Giday M, Asfaw Z, Woldu Z (2009) Medicinal plants of the Meinit ethnic group of Ethiopia:
an ethnobotanical study. J Ethnopharmacol 124:513–521
40. Girnun GD, Smith WM, Drori S, Sarraf P, Mueller E, Eng C, Nambiar P, Rosenberg DW,
Bronson RT, Edelmann W (2002) APC-dependent suppression of colon carcinogenesis by
PPARc. Proc Natl Acad Sci 99:13771–13776
41. Golan-Goldhirsh A, Gopas J (2013) Plant derived inhibitors of NF-jB. Phytochem Rev
13:107–121
42. Gottlieb TM, Leal JF, Seger R, Taya Y, Oren M (2002) Cross-talk between Akt, p53 and
Mdm2: possible implications for the regulation of apoptosis. Oncogene 21:1299–1303
43. Gp C (2012) Anthelmintic activity of fruits of Embelia ribes Burm. Int J Pharm Chem Sci
1:1680–1681
16 Embelin and Its Role in Chronic Diseases 413
44. Gustafson DL, Swanson JD, Pritsos CA (1993) Modulation of glutathione and glutathione
dependent antioxidant enzymes in mouse heart following doxorubicin therapy. Free Radic
Res Commun 19:111–120
45. Harish GU, Danapur V, Jain R, Patell VM (2012) Endangered medicinal plant Embelia ribes
Burm.f.—a review. Pharmacognosy Journal 4:6–19
46. Hendrickson BA, Ranjana G, Cho JH (2002) Clinical aspects and pathophysiology of
inflammatory bowel disease. Clin Microbiol Rev 15:79–94
47. Heo JY, Kim HJ, Kim SM, Park KR, Park SY, Kim SW, Nam D, Jang HJ, Lee SG, Ahn KS,
Kim SH, Shim BS, Choi SH, Ahn KS (2011) Embelin suppresses STAT3 signaling,
proliferation, and survival of multiple myeloma via the protein tyrosine phosphatase PTEN.
Cancer Lett 308:71–80
48. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM (1996)
IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-a-and
obesity-induced insulin resistance. Science 271:665–670
49. Hu R, Yang Y, Liu Z, Jiang H, Zhu K, Li J, Xu W (2015) The XIAP inhibitor Embelin
enhances TRAIL-induced apoptosis in human leukemia cells by DR4 and DR5 upregulation.
Tumour Biol 36:769–777
50. Hu R, Zhu K, Li Y, Yao K, Zhang R, Wang H, Yang W, Liu Z (2011) Embelin induces
apoptosis through down-regulation of XIAP in human leukemia cells. Med Oncol 28:1584–
1588
51. Huang Y, Lu J, Gao X, Li J, Zhao W, Sun M, Stolz DB, Venkataramanan R, Rohan LC, Li S
(2012) PEG-derivatized embelin as a dual functional carrier for the delivery of paclitaxel.
Bioconjug Chem 23:1443–1451
52. Issazadeh S, Mustafa M, Ljungdahl Å, Höjeberg B, Dagerlind Å, Elde R, Olsson T (1995)
Interferon c, interleukin 4 and transforming growth factor b in experimental autoimmune
encephalomyelitis in Lewis rats: dynamics of cellular mRNA expression in the central
nervous system and lymphoid cells. J Neurosci Res 40:579–590
53. Ivan L, Mehran M, Jean-Daniel C, Luc-Marie J, Christian S, Bénédicte B, Alain C, Alain R,
Pierre C, Simon W (2002) Reversible myocardial dysfunction in survivors of out-of-hospital
cardiac arrest. J Am Coll Cardiol 40:2110–2116
54. Jeon Y-J, Kim B-H, Kim S, Oh I, Lee S, Shin J, Kim T-Y (2013) Rhododendrin ameliorates
skin inflammation through inhibition of NF-jB, MAPK, and PI3K/Akt signaling. Eur J
Pharmacol 714:7–14
55. Johri R, Dhar S, Pahwa G, Sharma S, Kaul J, Zutshi U (1990) Toxicity studies with
potassium embelate, a new analgesic compound. Indian J Exp Biol 28:213–217
56. Jope RS, Johnson GVW (2004) The glamour and gloom of glycogen synthase kinase-3.
Trends Biochem Sci 29:95–102
57. Joshi R, Kamat JP, Mukherjee T (2007) Free radical scavenging reactions and antioxidant
activity of embelin: biochemical and pulse radiolytic studies. Chem Biol Interact 167:125–
134
58. Joy B, Kumar SN, Radhika AR, Abraham A (2015) Embelin (2,5-Dihydroxy-
3-undecyl-p-benzoquinone) for photodynamic therapy: study of their cytotoxicity in cancer
cells. Appl Biochem Biotechnol 175:1069–1079
59. Joy B, Nishanth Kumar S, Soumya MS, Radhika AR, Vibin M, Abraham A (2014) Embelin
(2,5-dihydroxy-3-undecyl-p-benzoquinone): a bioactive molecule isolated from Embelia
ribes as an effective photodynamic therapeutic candidate against tumor in vivo.
Phytomedicine 21:1292–1297
60. Kalyan Kumar G, Dhamotharan R, Kulkarni NM, Mahat MY, Gunasekaran J, Ashfaque M
(2011) Embelin reduces cutaneous TNF-alpha level and ameliorates skin edema in acute and
chronic model of skin inflammation in mice. Eur J Pharmacol 662:63–69
61. Kane LP, Smith SV, Stokoe D, Weiss A (1999) Induction of NF-kappaB by the Akt/PKB
kinase. Curr Biol 9:601–604
62. Karin M, Greten FR (2005) NF-jB: linking inflammation and immunity to cancer
development and progression. Nat Rev Immunol 5:749–759
414 H. Lu et al.
63. Karin M, Lin A (2002) NF-jB at the crossroads of life and death. Nat Immunol 3:221–227
64. Khan MI, Ahmed A, Akram M, Mohiuddin E, Khan U, Ayaz S, Shah S, Asif M, Ghazala S,
Ahmed K (2010) Monograph of Embelia ribes Burm. F. Afr J Plant Sci 4:503–505
65. Kim SW, Kim SM, Bae H, Nam D, Lee JH, Lee SG, Shim BS, Kim SH, Ahn KS, Choi SH,
Sethi G, Ahn KS (2013) Embelin inhibits growth and induces apoptosis through the
suppression of Akt/mTOR/S6K1 signaling cascades. Prostate 73:296–305
66. Krishnamoorthy G, Wekerle H (2009) EAE: an immunologist’s magic eye. Eur J Immunol
39(8):2031–2035
67. Kumar D, Kumar A, Prakash O (2012) Potential antifertility agents from plants: a
comprehensive review. J Ethnopharmacol 140:1–32
68. Kumar GK, Dhamotharan R, Kulkarni NM, Honnegowda S, Murugesan S (2011) Embelin
ameliorates dextran sodium sulfate-induced colitis in mice. Int Immunopharmacol 11:724–
731
69. Lamblin M, Sallustrau A, Commandeur C, Cresteil T, Felpin F-X, Dessolin J (2012)
Synthesis and biological evaluation of hydrophilic embelin derivatives. Tetrahedron
68:4655–4663
70. Lewis K (2001) Riddle of biofilm resistance. Antimicrob Agents Chemother 45:999–1007
71. Li M, Wang X, Meintzer MK, Laessig T, Birnbaum MJ, Heidenreich KA (2000)
Cyclic AMP promotes neuronal survival by phosphorylation of glycogen synthase kinase 3b.
Mol Cell Biol 20:9356–9363
72. Li Y, Sarkar FH (2002) Inhibition of nuclear factor jB activation in PC3 cells by genistein is
mediated via Akt signaling pathway. Clin Cancer Res 8:2369–2377
73. Liang Q, Yun D, Gu Q, Chan KW, Bing Z, Ma J, Wang J, Lan HY, Wong BCY (2008)
Down-regulation of X-linked inhibitor of apoptosis synergistically enhanced peroxisome
proliferator-activated receptor c ligand-induced growth inhibition in colon cancer. Mol
Cancer Ther 7:2203–2211
74. Liu A, Liu Y, Li PK, Li C, Lin J (2011) LLL12 inhibits endogenous and exogenous
interleukin-6-induced STAT3 phosphorylation in human pancreatic cancer cells. Anticancer
Res 31:2029–2035
75. Lopiccolo J, Blumenthal GM, Bernstein WB, Dennis PA (2008) Targeting the
PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist
Updates 11:32–50
76. Lu J, Huang Y, Zhao W, Marquez RT, Meng X, Li J, Gao X, Venkataramanan R, Wang Z,
Li S (2013) PEG-derivatized embelin as a nanomicellar carrier for delivery of paclitaxel to
breast and prostate cancers. Biomaterials 34:1591–1600
77. Mahendran S, Badami S, Maithili V (2011) Evaluation of antidiabetic effect of embelin from
Embelia ribes in alloxan induced diabetes in rats. Biomed Prev Nutr 1:25–31
78. Mahendran S, Thippeswamy BS, Veerapur VP, Badami S (2011) Anticonvulsant activity of
embelin isolated from Embelia ribes. Phytomedicine 18:186–188
79. Makawiti DW, Konji VN, Olowookere JO (1990) Interaction of benzoquinones with
mitochondria interferes with oxidative phosphorylation characteristics. FEBS Lett 266:26–28
80. Manna S, Bhattacharyya D, Basak D, Mandal T (2004) Single oral dose toxicity study of
a-cypermethrin in rats. Indian J Pharmacol 36:25
81. Mantovani A, Allavena P, Sica A, Balkwill F (2008) Cancer-related inflammation. Nature
454:436–444
82. Mcauliffe PF, Meric-Bernstam F, Mills GB, Gonzalez-Angulo AM (2010) Deciphering the
role of PI3K/Akt/mTOR pathway in breast cancer biology and pathogenesis. Clin Breast
Cancer 10:S59–S65
83. Mencher SK, Wang LG (2005) Promiscuous drugs compared to selective drugs (promiscuity
can be a virtue). BMC Clin Pharmacol 5:3
84. Mj VDV (2004) Overexpression of P70 S6 kinase protein is associated with increased risk of
locoregional recurrence in node-negative premenopausal early breast cancer patients. Br J
Cancer 90:1543–1550
16 Embelin and Its Role in Chronic Diseases 415
85. Moller DE (2000) Potential role of TNF-alpha in the pathogenesis of insulin resistance and
type 2 diabetes. Trends Endocrinol Metab TEM 11:212–217
86. Mori T, Doi R, Kida A, Nagai K, Kami K, Ito D, Toyoda E, Kawaguchi Y, Uemoto S (2007)
Effect of the XIAP inhibitor embelin on TRAIL-induced apoptosis of pancreatic cancer cells.
J Surg Res 142:281–286
87. Muris JJF, Cillessen SAGM, Wim V, Houdt IS, Kummer JA, Van Krieken JHJM, Jiwa NM,
Jansen PM, Kluin-Nelemans HC, Van Ossenkoppele GJ (2005) Immunohistochemical
profiling of caspase signaling pathways predicts clinical response to chemotherapy in
primary nodal diffuse large B-cell lymphomas. Blood 105:2916–2923
88. Naik SR, Niture NT, Ansari AA, Shah PD (2013) Anti-diabetic activity of embelin:
involvement of cellular inflammatory mediators, oxidative stress and other biomarkers.
Phytomedicine 20:797–804
89. Narayanaswamy R, Gnanamani A (2014) 2, 5-dihydroxy-3-undecyl-1, 4-benzoquinone
(Embelin)-a second solid gold of India- A review. Int J Pharm Pharm Sci 6:23–30
90. Narayanaswamy R, Shymatak M, Chatterjee S, Wai LK, Arumugam G (2014) Inhibition of
angiogenesis and nitric oxide synthase (NOS), by Embelin & Vilangin using in vitro, in vivo
and in silico STUDIES. Adv Pharm Bull 4:543
91. Newton R, Solomon K, Covington M, Decicco C, Haley P, Friedman S, Vaddi K (2001)
Biology of TACE inhibition. Ann Rheum Dis 60:iii25–iii32
92. Nikolovska-Coleska Z, Xu L, Hu Z, Tomita Y, Li P, Roller PP, Wang R, Fang X, Guo R,
Zhang M, Lippman ME, Yang D, Wang S (2004) Discovery of embelin as a cell-permeable,
small-molecular weight inhibitor of XIAP through structure-based computational screening
of a traditional herbal medicine three-dimensional structure database. J Med Chem 47:2430–
2440
93. Oh K, Lee O-Y, Shon SY, Nam O, Ryu PM, Seo MW, Lee D-S (2013) A mutual activation
loop between breast cancer cells and myeloid-derived suppressor cells facilitates spontaneous
metastasis through IL-6 trans-signaling in a murine model. Breast Cancer Res 15:R79
94. Park N, Baek HS, Chun Y-J (2015) Embelin-induced apoptosis of human prostate cancer
cells is mediated through modulation of Akt and b-catenin signaling. PLoS ONE 10:
e0134760
95. Parmar K, Patel J, Sheth N (2014) Fabrication and characterization of liquisolid compacts of
Embelin for dissolution enhancement. J Pharm Invest 44:391–398
96. Parmar K, Patel J, Sheth N (2015) Self nano-emulsifying drug delivery system for Embelin:
design, characterization and in-vitro studies. Asian J Pharm Sci. http://www.elsevier.com/
locate/AJPS
97. Patel RK, Patel VR, Patel MG (2012) Development and validation of a RP-HPLC method for
the simultaneous determination of embelin, rottlerin and ellagic acid in Vidangadi churna.
J Pharm Anal 2:366–371
98. Pathan RA, Bhandari U (2010) Preparation & characterization of embelin–phospholipid
complex as effective drug delivery tool. J Incl Phenom Macrocycl Chem 69:139–147
99. Peng M, Huang B, Zhang Q, Fu S, Wang D, Cheng X, Wu X, Xue Z, Zhang L, Zhang D,
Da Y, Dai Y, Yang Q, Yao Z, Qiao L, Zhang R (2014) Embelin inhibits pancreatic cancer
progression by directly inducing cancer cell apoptosis and indirectly restricting IL-6
associated inflammatory and immune suppressive cells. Cancer Lett 354:407–416
100. Peng M, Xu S, Zhang Y, Zhang L, Huang B, Fu S, Xue Z, Da Y, Dai Y, Qiao L, Dong A,
Zhang R, Meng W (2014) Thermosensitive injectable hydrogel enhances the antitumor effect
of embelin in mouse hepatocellular carcinoma. J Pharm Sci 103:965–973
101. Perkins ND (2012) The diverse and complex roles of NF-jB subunits in cancer. Nat Rev
Cancer 12:121–132
102. Phuong do T, Ma CM, Hattori M, Jin JS (2009) Inhibitory effects of antrodins A-E from
Antrodia cinnamomea and their metabolites on hepatitis C virus protease. Phytother Res
23:582–584
103. Plutzky J (2003) PPARs as therapeutic targets: Reverse cardiology? Science 302:406–407
416 H. Lu et al.
104. Podolak I, Galanty A, Janeczko Z (2005) Cytotoxic activity of embelin from Lysimachia
punctata. Fitoterapia 76:333–335
105. Podolak I, Strzałka M (2008) Qualitative and quantitative LC profile of embelin and
rapanone in selected Lysimachia species. Chromatographia 67:471–475
106. Poojari R (2014) Embelin—a drug of antiquity: shifting the paradigm towards modern
medicine. Expert Opin Investig Drugs 23:427–444
107. Poojari RJ (2014) Embelin, a small molecule quinone with a co-clinical power for
castrate-resistant prostate cancer. Front Pharmacol 5:184
108. Radhakrishnan N (2011) A potential antibacterial agent Embelin, a natural benzoquinone
extracted from Embelia ribes. Biol Med 3(2):1–7
109. Rani P, Khullar N (2004) Antimicrobial evaluation of some medicinal plants for their
anti-enteric potential against multi-drug resistant Salmonella typhi. Phytother Res 18:670–
673
110. Rathi SG, Bhaskar VH, Patel PG (2010) Antifungal activity of Embelia ribes plant extracts.
Int J Pharm Biol Res 1:6–10
111. Rathinam KKS, Ramiah N (1976) Studies on the antifertility activity of embelin. J Res Ind
Med Yoga Homeop 11:84–90
112. Reuter S, Prasad S, Phromnoi K, Kannappan R, Yadav VR, Aggarwal BB (2010) Embelin
suppresses osteoclastogenesis induced by receptor activator of NF-jB ligand and tumor cells
in vitro through inhibition of the NF-jB cell signaling pathway. Mol Cancer Res 8:1425–
1436
113. Rhodus NLCB, Myers S, Miller L, Ho V, Ondrey F (2005) The feasibility of monitoring
NF-jB associated cytokines: TNF-alpha, IL-1alpha, IL-6, and IL-8 in whole saliva for the
malignant transformation of oral lichen planus. Mol Carcinog 44:77–82
114. Riedl S, Renatus M, Schwarzenbacher R, Zhou Q, Sun C, Fesik S, Liddington R, Salvesen G
(2001) Structural basis for the inhibition of caspase-3 by XIAP. Cell 104:791–800
115. Rondevaldova J, Leuner O, Teka A, Lulekal E, Havlik J, Van Damme P, Kokoska L (2015)
In vitro antistaphylococcal effects of Embelia schimperi extracts and their component
embelin with oxacillin and tetracycline. Evid Based Complement Alternat Med 2015:1–7
(ID: 175983)
116. Ruiz-Bailén M, Hoyos EAD, Ruiz-Navarro S, Díaz-Castellanos MÁ, Rucabado-Aguilar L,
Gómez-Jiménez FJ, Martínez-Escobar S, Moreno RM, Fierro-Rosón J (2005) Reversible
myocardial dysfunction after cardiopulmonary resuscitation. Resuscitation 66:175–181
117. Sahu AK, Gautam MK, Deshmukh PT, Kushwah LS, Silawat N, Akbar Z, Muthu MS (2012)
Effect of embelin on lithium–induced nephrogenic diabetes insipidus in albino rats. Asian
Pac J Trop Dis 2:S729–S733
118. Sahu BD, Anubolu H, Koneru M, Kumar JM, Kuncha M, Rachamalla SS, Sistla R (2014)
Cardioprotective effect of embelin on isoproterenol-induced myocardial injury in rats:
possible involvement of mitochondrial dysfunction and apoptosis. Life Sci 107:59–67
119. Salvesen GS, Duckett CS (2002) IAP proteins: blocking the road to death’s door. Nat Rev
Mol Cell Biol 3:401–410
120. Samatha S, Vasudevan TN (1996) Embelia ribes Burm. as a source of red colorant. J Sci Ind
Res 55:888–889
121. Sandro A, Gabriele BP (2005) Biologic therapy for inflammatory bowel disease. Drugs
65:2253–2286
122. Schaible AM, Traber H, Temml V, Noha SM, Filosa R, Peduto A, Weinigel C, Barz D,
Schuster D, Werz O (2013) Potent inhibition of human 5-lipoxygenase and microsomal
prostaglandin E2 synthase-1 by the anti-carcinogenic and anti-inflammatory agent embelin.
Biochem Pharmacol 86:476–486
123. Schreiner SJ, Schiavone AP, Smithgall TE (2002) Activation of STAT3 by the Src family
kinase Hck requires a functional SH3 domain. J Biol Chem 277:45680–45687
124. Shankarmurthy K, Krishna V, Maruthi K, Rahiman B (2004) Rapid adventitious
organogenesis from leaf segments of Embelia ribes Burm.-a threatened medicinal plant.
TAIWANIA-TAIPEI- 49:194–200
16 Embelin and Its Role in Chronic Diseases 417
125. Shelar RMC, Tekale P, Katkar K, Naik V, Suthar A, Chauhan VS (2009) Embelin: an HPLC
method for quantitative estimation in Embelia ribes Burm. F. Int J Pharm Clin Res 1:146–
149
126. Shi Y (2009) Structural biology of programmed cell death. In: Essentials of apoptosis.
Springer, pp. 95–118
127. Shimada T, Kojima K, Yoshiura K, Hiraishi H, Terano A (2002) Characteristics of the
peroxisome proliferator activated receptor c (PPARc) ligand induced apoptosis in colon
cancer cells. Gut 50:658–664
128. Singh B, Guru SK, Sharma R, Bharate SS, Khan IA, Bhushan S, Bharate SB,
Vishwakarma RA (2014) Synthesis and anti-proliferative activities of new derivatives of
embelin. Bioorg Med Chem Lett 24:4865–4870
129. Singh D, Singh R, Singh P, Gupta RS (2009) Effects of embelin on lipid peroxidation and
free radical scavenging activity against liver damage in rats. Basic Clin Pharmacol Toxicol
105:243–248
130. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer DD, Wang H-G,
Reed JC, Nicholson DW, Alnemri ES (1999) Ordering the cytochrome c–initiated caspase
cascade: hierarchical activation of caspases-2,-3,-6,-7,-8, and-10 in a caspase-9–dependent
manner. J Cell Biol 144:281–292
131. Sreepriya M, Bali G (2005) Chemopreventive effects of embelin and curcumin against
N-nitrosodiethylamine/phenobarbital-induced hepatocarcinogenesis in Wistar rats.
Fitoterapia 76:549–555
132. Sreepriya M, Bali G (2006) Effects of administration of Embelin and Curcumin on lipid
peroxidation, hepatic glutathione antioxidant defense and hematopoietic system during
N-nitrosodiethylamine/Phenobarbital-induced hepatocarcinogenesis in Wistar rats. Mol Cell
Biochem 284:49–55
133. Sumalatha KR, Abiramasundari G, Chetan GK, Divya T, Sudhandiran G, Sreepriya M
(2014) XIAP inhibitor and antiestrogen embelin abrogates metastasis and augments
apoptosis in estrogen receptor positive human breast adenocarcinoma cell line MCF-7.
Mol Biol Rep 41:935–946
134. Suthar M, Patel R, Hapani K, Patel A (2009) Screening of Embelia ribes for antifungal
activity. Int J Pharm Sci Drug Res 1:203–206
135. Tambekar DH, Khante BS, Chandak BR, Titare AS, Boralkar SS, Aghadte SN (2009)
Screening of antibacterial potentials of some medicinal plants from Melghat forest in India.
Afr J Tradit Complement Altern Med 6:228–232
136. Tamm I, Kornblau SM, Segall H, Krajewski S, Welsh K, Kitada S, Scudiero DA, Tudor G,
Qui YH, Monks A, Andreeff M, Reed JC (2000) Expression and prognostic significance of
IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res 6:1796–1803
137. Thippeswamy BS, Mahendran S, Biradar MI, Raj P, Srivastava K, Badami S, Veerapur VP
(2011) Protective effect of embelin against acetic acid induced ulcerative colitis in rats. Eur J
Pharmacol 654:100–105
138. Thippeswamy BS, Nagakannan P, Shivasharan BD, Mahendran S, Veerapur VP, Badami S
(2011) Protective effect of embelin from Embelia ribes Burm. against transient global
ischemia-induced brain damage in rats. Neurotox Res 20:379–386
139. Varier PS (2006) Indian medicinal plants a compendium of 500 species, vol 2. Orient
Longman (Pvt) Ltd., Chennai, pp 368–371
140. Vinten-Johansen J, Jiang R, Reeves JG, Mykytenko J, Deneve J, Jobe LJ (2007)
Inflammation, proinflammatory mediators and myocardial ischemia-reperfusion Injury.
Hematol Oncol Clin North Am 21:123–145
141. Wang DG, Sun YB, Ye F, Li W, Kharbuja P, Gao L, Zhang DY, Suo J (2014) Anti-tumor
activity of the X-linked inhibitor of apoptosis (XIAP) inhibitor embelin in gastric cancer
cells. Mol Cell Biochem 386:143–152
142. Wang L, Yi TM (2009) IL-17 can promote tumor growth through an IL-6-Stat3 signaling
pathway. J Exp Med 206:1431–1438
418 H. Lu et al.
143. Watson RT, Makoto K, Pessin JE (2004) Regulated membrane trafficking of the
insulin-responsive glucose transporter 4 in adipocytes. Endocr Rev 25:177–204
144. Xue Z, Ge Z, Zhang K, Sun R, Yang J, Han R, Peng M, Li Y, Li W, Zhang D, Hao J, Da Y,
Yao Z, Zhang R (2014) Embelin suppresses dendritic cell functions and limits autoimmune
encephalomyelitis through the TGF-beta/beta-catenin and STAT3 signaling pathways. Mol
Neurobiol 49:1087–1101
145. Yang T, Lan J, Huang Q, Chen X, Sun X, Liu X, Yang P, Jin T, Wang S, Mou X (2015)
Embelin sensitizes acute myeloid leukemia cells to TRAIL through XIAP inhibition and
NF-kappaB inactivation. Cell Biochem Biophys 71:291–297
146. Yasmina L, Terrence T, David J, Elise T, Yisong W, Kuchroo VK, Flavell RA (2008) TGF-b
signaling in dendritic cells is a prerequisite for the control of autoimmune encephalomyelitis.
Proc Natl Acad Sci USA 105:10865–10870
147. Yen D, Cheung J, Scheerens H, Poulet F, McClanahan T, McKenzie B, Kleinschek MA,
Owyang A, Mattson J, Blumenschein W, Murphy E, Sathe M, Cua DJ, Kastelein RA,
Rennick D (2006) IL-23 is essential for T cell-mediated colitis and promotes inflammation
via IL-17 and IL-6. J Clin Invest 116:1310–1316
148. Yoshizumi T, Ohta TI, Terada I, Fushida S, Fujimura T, Nishimura G, Shimizu K, Yi S,
Miwa K (2004) Thiazolidinedione, a peroxisome proliferator-activated receptor-gamma
ligand, inhibits growth and metastasis of HT-29 human colon cancer cells through
differentiation-promoting effects. Int J Oncol 25:631–639
149. Zhao ZG, Tang ZZ, Zhang WK, Li JG (2015) Protective effects of embelin on myocardial
ischemia-reperfusion injury following cardiac arrest in a rabbit model. Inflammation 38:527–
533
150. Zou B, Qiao L, Wong BC (2009) Current understanding of the role of PPARgamma in
gastrointestinal cancers. PPAR Res 2009(1):121-132. (ID: 816957)
Chapter 17
Butein and Its Role in Chronic Diseases
Abstract Natural compounds isolated from various plant sources have been used
for therapeutic purpose for centuries. These compounds have been routinely used
for the management of various chronic ailments and have gained considerable
attention because of their significant efficacy and comparatively low side effects.
Butein, a chacolnoid compound that has been isolated from various medicinal
plants has exhibited a wide range of beneficial pharmacological effects, such as
anti-inflammatory, anticancer, antioxidant, and anti-angiogenic in diverse disease
models. This article briefly summarizes the past published literature related to the
therapeutic and protective effects of butein, as demonstrated in various models of
human chronic diseases. Further analysis of its important cellular targets, toxicity,
and pharmacokinetic profile may further significantly expand its therapeutic
application.
Z. Song H. Yu
Department of Physiology, Yong Loo Lin School of Medicine, MD9-04-11,
2 Medical Drive, Singapore 117597, Singapore
M.K. Shanmugam G. Sethi (&)
Department of Pharmacology, Yong Loo Lin School of Medicine,
National University of Singapore, Singapore 117600, Singapore
e-mail: [email protected]
H. Yu
Institute of Biotechnology and Nanotechnology, A*STAR, The Nanos,
#04-01, 31 Biopolis Way, Singapore 138669, Singapore
H. Yu
Singapore-MIT Alliance for Research and Technology, 1 CREATE Way,
#10-01 CREATE Tower, Singapore 138602, Singapore
H. Yu
Mechanobiology Institute, National University of Singapore, T-Lab, #05-01,
5A Engineering Drive 1, Singapore 117411, Singapore
17.1 Introduction
Plant-derived natural compounds and their derivatives have been used as thera-
peutic agents over the centuries. However, their exact chemical structures, functions
as well as molecular targets are not completely known as of yet. Their high ther-
apeutic efficiency and low side effects have made themselves ideal alternatives of
synthetic drugs [4, 56]. Thus extensive qualitative and quantitative tests have been
done to evaluate and to extend their therapeutic potential as drug candidates.
Butein, a plant polyphenol (2′,3,4,4′-2′,4′,3,4- or 3,4,2′,4′-tetrahydroxychalcone),
with the chemical structure shown in Fig. 17.1, has been reported to exhibit several
important pharmacological effects, such as anti-inflammatory, anti-cancer,
anti-oxidant, and anti-angiogenic [5, 33, 37, 50, 58]. It was first found in
Toxicodendronvernicifluum (formerly called RhusVerniciflua Stokes, RVS), a tree
widely used as a local food additive and therapeutic supplement in South East Asia
[62]. It can also be isolated from the heartwood of Dalbergiaodorifera, the seed of
Cyclopiasubternata and the stems of Semecarpusanacardium, as well as many
other plants [41]. The various plant sources of butein are shown in Table 17.1 [60].
The first dietary butein supplement Inh-AR, launched by Megabol, is a sport
pro-hormone supplement to control estrogen production by inhibiting aromatase
[1]. Its activities in aromatase inhibition and estrogen regulation have also made
butein a promising candidate for breast cancer treatment. Besides cancer [2, 30, 73],
it also has exhibited promising effects in the treatment of many other chronic
diseases, including inflammation [63], glaucoma [14], cardiovascular diseases [9],
and metabolic complications [48]. Although molecular mechanisms underlying its
various reported pharmacological effects remain to be elucidated, butein has been
found to modulate several important cellular signaling pathways involved in eti-
ology of chronic diseases. Meanwhile, it exhibits low toxicity against normal cells,
thereby indicating its potential both as a chemopreventive and chemotherapeutic
agent [28, 71]. In this chapter, we have briefly summarized various published
reports related to the pharmacological properties and biological activities of butein,
and emphasized on its therapeutic effects against various chronic diseases.
As a multi-targeted compound with diverse biological effects, butein has been reported
to target several important cell signaling pathways. Although the underlying mecha-
nisms and molecular targets remain to be elucidated, published results indicate that
butein exhibit therapeutic effects on several chronic diseases including inflammation,
cancer, cardiovascular diseases, and metabolic complications. This section briefly
summarizes its reported therapeutic effects against various chronic ailments.
17.3.2 Cancer
As stated above, butein can interfere with tumor progression by suppressing inflam-
matory reactions [52]. It is also well recognized as a promising anticancer agent and
functions by inhibiting cellular growth, migration as well as by promoting apoptosis. Its
anticancer effects have been reported against various tumor cells, including breast
cancer [67], colon cancer [74], leukemia [31], melanoma [15], osteosarcoma cells [24],
prostate cancer [30], hepatocellular carcinoma [57], lung cancer [42], and neuroblas-
toma cells [8]. Its anticancer effects are achieved by modulating various critical cellular
signaling pathways and enzymes involved in tumorigenesis and apoptosis. It is well
established that PI3K/Akt/mTOR is one of the major signaling pathways regulating cell
survival and motility [17]. And its activation plays a key role in tumor cell growth and
invasion [6, 17]. Butein can significantly suppress PI3K/Akt phosphorylation in human
prostate cancer cells [30] and cervical cancer cells [2]. It is also found to downregulate
MMP-9 and uPA expression by inhibiting Akt/mTOR/p70S6K, thus acting as anti-
metastatic agent [46].
Various studies also show that butein induces reactive oxygen species
(ROS) production, which is also critical for cell apoptosis, cell senescence and cell
division [65]. It has been reported that it induces G2/M phase cell cycle arrest in
hepatoma cells by regulating ROS level, JNK expression, ATM, and Chk activity
[50]. It also induces ROS generation, ERK1/2, and p38MAPK suppression in
triple-negative breast cancer MDA-MB-231 cells [72]. Similarly, it causes apop-
tosis of neutroblastoma cells by increasing ROS level and modulating various
enzymes [8]. And it also shows antiproliferative and apoptotic activities in HeLa
human cervical cancer cells, by inhibiting the expression of PI3K, Akt and mTOR
phosphorylation, and by inducing the ROS generation [2]. These effects were
further confirmed in vivo in a nude mouse model, where it significantly suppressed
cervical tumor growth with limited side effects by inhibiting PI3K/Akt/mTOR
activation and increasing ROS generation [2]. It also increases caspase-3, -8, -9
activities in neutroblastoma cells and caspase-3 activity in HeLa cells, the
expression of which is critical for apoptosis [31]. Moreover, Bcl-2 downregulation
is also observed in butein-induced apoptosis of neutroblastoma cells [8]. It is also
shown that butein increases Bax while suppressing Bcl-2 expression in Hela cells.
Bax and Bcl-2 are both critical enzymes for apoptosis, and the Bax/Bcl-2 ratio
contributes to the butein-induced apoptosis [2].
Another critical molecule involved in the anticancer activity of butein is the
signal transducer and activator of transcription 3 (STAT3), a transcription factor.
STAT3 can regulate the NF-jB signaling pathway, and thus plays a major role in
the inflammation-related tumorigenesis [21, 76]. It is activated by IL6/JAK sig-
naling pathway [29]. The IL6/JAK/STAT3 mechanism has been found to partici-
pate in the progression of various malignant tumors, such as prostate cancer, breast
cancer, and myeloma [41]. Butein inhibits the IL6/JAK/STAT3 pathway by sup-
pressing c-Src and JAK1/JAK2 activation, and by inhibiting Bcl-xL, Bcl-2, and
cyclin D1 expression [57]. Moreover, it blocks the ERK1/2 and NF-jB signaling
17 Butein and Its Role in Chronic Diseases 425
pathways in human bladder cancer cells, which inhibits the cell proliferation and tumor
invasion [78]. It also negatively regulates NK-jB activation by modulating critical
molecules such as TNFa1, TRADD, TRAF2, NIK, TAK1/TAB17.1, and IKK-b [55].
Besides STAT3, matrix metalloproteinase (MMP) is also critical in tumor cell apop-
tosis, tumor progression, and invasion. It has been found that butein suppresses the
activation of MMP-9 in prostate cancer cells [18, 35], and the activation of MMP-2 and
MMP-9 in Hela cells [2]. Butein is also noted to inhibit both MMP-9 activity and
STAT3 activation in Colo 205 cells [41], which indicates its possible therapeutic
potential for treating bowel inflammation-induced colon cancer.
There are several critical enzymes and molecules regulated by butein in various
tumor cell lines. For example, glutathione-s -transferase (GST), an enzyme that
reduces H2O2-induced oxidative stress, is significantly increased after butein
treatment in human caucasian neuroblastoma IMR-32 cells [77]. And it also sup-
presses some other enzymes critical for tumorigenesis, such as histone deacetylase
enzymes [54], aromatase [67] and recombinant human aldo-keto reductase family
[61]. And butein can inhibit DNA, RNA, and protein synthesis by reducing the
cellular uptake of thymidine, uridine, and leucine of cancer cells [74]. Moreover,
butein treatment suppresses colony formation in UACC-812 human breast cancer
cells co-cultured with fibroblasts [34]. And it induces apoptosis in U937 human
leukemia cells by modulating caspase 3-dependent pathways [23].
Cardiovascular diseases are one of the leading causes of mortality worldwide [49].
They are also closely associated with various other complications, such as obesity
and diabetes [20]. Butein has been found to exhibit cardiovascular protection effects
by improving blood circulation, in which platelets aggregation and thrombosis play
a key role. It has been reported that butein also exhibits anti-thrombin effects by
inhibiting the blood coagulation drawn from New Zealand white rabbits [43]. It also
inhibits ADP-induced platelet aggregation [43] and adrenalin-induced secondary
aggregation in human plasma [45]. Besides its anti-platelet effects, butein can also
suppress the viability and motility of vascular smooth muscle cells, both of which
can lead to atherosclerosis by thickening the blood vessels [9]. This activity is
further confirmed in vivo in animal models of phenylephrine-induced contracted rat
aorta [75] and balloon-injured rat carotid arteries [8].
streptozotocin [44]. It also has been found to have protective effects on functional
pancreatic beta-cells and rat islets. Type I diabetes is mainly caused by beta-cell
destruction, in which various inflammatory cytokines are involved, including IL-1b,
TNF-a, IFN-c, etc. [25]. Moreover, it protects the cellular activity of beta cells by
inhibiting cytokine-mediated cell death, NO production, iNOS expression, and
NF-jB translocation [25], thus indicating its potential for Type I diabetes mellitus
prevention.
It has been reported that butein is also active in liver fibrosis prevention, by
increasing albumin production [16] and inhibiting hepatic stellate cell proliferation
[69]. In vivo data shows that butein can reduce hydroxyproline and malondialde-
hyde levels in liver fibrosis mouse model induced by CCl4 [36]. It can also decrease
the toxicity caused by ethanol in hepatic stellate cells and hepatoma HepG2 cells
[64], indicating its potential to prevent alcohol-related liver complications, such as
cirrhosis, fatty liver, and alcoholic hepatitis. The effects to decrease alcohol-induced
toxicity are achieved by suppressing ROS production, MMP-1 and -2 inhibitors in
HSCs. It has been also found that butein inhibits the JNK/p38 MAPK signaling
pathway and upregulates NF-jB [64]. Moreover, butein inhibits rat hepatocyte
apoptosis by decreasing PARP cleavage, DNA fragmentation, and caspase acti-
vation [60].
Butein has been found to have therapeutic effects against the initiation and pro-
gression of several other major human diseases. For example, it has anti-HIV effects
by causing around 58 % inhibition of HIV-1 protease [70]. It can also suppress
ICAM-1 expression and increase leukocyte function-associated antigen-1 positive
cells in nephritic glomeruli [19]. And it can decreases the protein level in urine and
cholesterol level in plasma, thus indicating its therapeutic potential for nephropathy,
a kidney disease which can be caused by either IgA deposition in glomerulus or
chemotherapy agents [60]. Moreover, it is potential drug candidate for tuberculosis,
as it can inhibit myocolic acids production and HadB activity, by blocking the
activity of Mycobacteriumbovis BCG, a myobacteria that causes tuberculosis [3]. It
also shows protective effects in several neuronal cell lines. It largely reduces the
toxicity induced by glutamate in mouse HT22 neuronal cells [11, 12]. And it can be
a potential pain killer by inhibiting SNARE-complex, a critical enzyme involved in
the synaptic vesicle transmission [27].
17 Butein and Its Role in Chronic Diseases 427
17.5 Conclusions
Herbal medicine can be traced back to hundreds of years ago, and our ancestors use
different plants with different bioactivities in different combinations. However, the
ancient application of herbal medicine is solely based on experience, without
qualitative or quantitative tests. With the development of drug testing techniques,
the chemical structures and activities of these therapeutic compounds have been
extensively studied, by using high throughput screening and various in vitro as well
as in vivo models. Butein is a multi-target compound that can exhibit a wide array
of beneficial effects by modulating a wide variety of cellular signal transduction
428 Z. Song et al.
TNF-α
PI3K/Akt/mTOR
ROS
Butein
c-Src
ß-glucuronidase
Fig. 17.2 Major signaling pathways and enzymes regulated by butein, # indicates inhibition or
downregulation; " indicates activation or upregulation. The molecules and pathways in the inner
circle are all upregulated or activated, while those in the outer two circles are downregulated or
inhibited by butein
cascades (Fig. 17.2). Its therapeutic effects have been validated in various cell lines
as well as mice models. However, neither Food and Drug Administration (FDA) in
US nor European Food Safety Authority (EFSA) has approved it as a therapeutic
drug. The only formulation containing butein available at present is Inh-AR, a
dietary supplement. There are many structure-activity studies on butein to reveal the
relationship between its chemical structural features and reported pharmacological
effects [22, 53, 63]. These studies provides in-depth understanding of its observed
biological effects and may also aid to synthesize similar compounds in future that
can achieve the same, or even better, therapeutic effects with better pharmacokinetic
profile. Although butein has many beneficial effects and low side effects, its toxicity
still needs to be elucidated. It is shown that the RhusVerniciflua Stokes, one of the
17 Butein and Its Role in Chronic Diseases 429
original sources of butein, can cause severe skin complications [32], thereby
indicating that butein may also have potential adverse effects. However, there is
currently lack of relevant toxicity studies with this therapeutic agent. Overall, butein
appears to be a promising drug candidate because of its various beneficial effects
and therapeutic actions against major human chronic diseases. However, additional
clinical trials are needed to validate its safety and efficacy so that it can be rapidly
applied for human use in near future.
Acknowledgments This work was supported by National University Health System Bench to
Bedside grant to GS.
References
14. Choi SJ, Lee MY, Jo H, Lim SS, Jung SH (2012) Preparative isolation and purification of
neuroprotective compounds from Rhus verniciflua by high speed counter-current
chromatography. Biol Pharm Bull 35(4):559–567
15. Cui Z, Song E, Hu DN, Chen M, Rosen R, McCormick SA (2012) Butein induces apoptosis in
human uveal melanoma cells through mitochondrial apoptosis pathway. Curr Eye Res 37
(8):730–739
16. Dong MS, Jung NC, Na CS (2004) Composition comprising phenolic compound for
preventing and treating liver cirrhosis. Patent no.: WO2004002471 A1
17. Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C, Gonzalez-Baron M (2004)
PI3K/Akt signalling pathway and cancer. Cancer Treat Rev 30(2):193–204
18. Garg P, Vijay-Kumar M, Wang L, Gewirtz AT, Merlin D, Sitaraman SV (2009) Matrix
metalloproteinase-9-mediated tissue injury overrides the protective effect of matrix
metalloproteinase-2 during colitis. Am J Physiol Gastrointest Liver Physiol 296(2):G175–
G184
19. Hayashi K, Nagamatsu T, Honda S, Suzuki Y (1996) Butein (3,4,2’,4’-tetrahydroxychalcone)
ameliorates experimental anti-glomerular basement membrane antibody-associated
glomerulonephritis (3). Eur J Pharmacol 316:297–306
20. Highlander P, Shaw GP (2010) Current pharmacotherapeutic concepts for the treatment of
cardiovascular disease in diabetics. Ther Adv Cardiovasc Dis 4(1):43–54
21. Hodge DR, Hurt EM, Farrar WL (2005) The role of IL-6 and STAT3 in inflammation and
cancer. Eur J Cancer 41(16):2502–2512
22. Ishikawa YT, Goto M, Yamaki K (2003) Inhibitory effects of several flavonoids on E-selectin
expression on human umbilical vein endothelial cells stimulated by tumor necrosis factor-a.
Phytother Res 17:1224–1227
23. Iwashita KK, Kobori M, Yamaki K, Tsushida T (2000) Flavonoids inhibit cell growth and in
B16 melanoma 4A5 Cells induce apoptosis. Biosci Biotechnol Biochem 64(9):1813–1820
24. Jang HS, Kook SH, Son YO, Kim JG, Jeon YM, Jang YS, Choi KC, Kim J, Han SK, Lee KY,
Park BK, Cho NP, Lee JC (2005) Flavonoids purified from Rhus verniciflua Stokes actively
inhibit cell growth and induce apoptosis in human osteosarcoma cells. Biochim Biophys Acta
1726(3):309–316
25. Jeong GS, Lee DS, Song MY, Park BH, Kang DG, Lee HS, Kwon KB, Kim YS (2011) Butein
from Rhus verniciflua protects pancreatic b cells against cytokine-induced toxicity mediated
by inhibition of nitric oxide formation. Biol Pharm Bull 34(1):97–102
26. Jung CH, Kim JH, Hong MH, Seog HM, Oh SH, Lee PJ, Kim GJ, Kim HM, Um JY, Ko SG
(2007) Phenolic-rich fraction from Rhus verniciflua Stokes (RVS) suppress inflammatory
response via NF-kappaB and JNK pathway in lipopolysaccharide-induced RAW 264.7
macrophages. J Ethnopharmacol 110(3):490–497
27. Jung CH, Kweon DH, Shin YK, Yang YS (2011) Polyphenol compounds with modulating
neurotransmitter release. Patent no.: US8263643 B2
28. Kang DG, Lee AS, Mun YJ, Woo WH, Kim YC, Sohn EJ, Moon MK, Lee HS (2004) Butein
ameliorates renal concentrating ability in cisplatin-induced acute renal failure in rats. Biol
Pharm Bull 27(3):366–370
29. Kato T (2011) Stat3-driven cancer-related inflammation as a key therapeutic target for cancer
immunotherapy. Immunotherapy 3(5):587–591
30. Khan N, Adhami VM, Afaq F, Mukhtar H (2012) Butein induces apoptosis and inhibits
prostate tumor growth in vitro and in vivo. Antioxid Redox Signal 16(11):1195–1204
31. Kim NY, Pae HO, Oh GS, Kang TH, Kim YC, Rhew HY, Chung HT (2001) Butein, a plant
polyphenol, induces apoptosis concomitant with increased caspase-3 activity, decreased Bcl-2
expression and increased Bax expression in HL-60 cells. Pharmacol Toxicol 88:261–266
32. Kim KH, Moon E, Choi SU, Pang C, Kim SY, Lee KR (2015) Identification of cytotoxic and
anti-inflammatory constituents from the bark of Toxicodendron vernicifluum (Stokes) F.A.
Barkley. J Ethnopharmacol 162:231–237
17 Butein and Its Role in Chronic Diseases 431
33. Kojima R, Kawachi M, Ito M (2015) Butein suppresses ICAM-1 expression through the
inhibition of IkappaBalpha and c-Jun phosphorylation in TNF-alpha- and PMA-treated
HUVECs. Int Immunopharmacol 24(2):267–275
34. Kook SH, Son YO, Chung SW, Lee SA, Kim JG, Jeon YM, Lee JC (2007)
Caspase-independent death of human osteosarcoma cells by flavonoids is driven by
p53-mediated mitochondrial stress and nuclear translocation of AIF and endonuclease G.
Apoptosis 12(7):1289–1298
35. Lakatos G, Sipos F, Miheller P, Hritz I, Varga MZ, Juhasz M, Molnar B, Tulassay Z,
Herszenyi L (2012) The behavior of matrix metalloproteinase-9 in lymphocytic colitis,
collagenous colitis and ulcerative colitis. Pathol Oncol Res 18(1):85–91
36. Lee SH, Nan JX, Zhao YZ, Woo SW, Park EJ, Kang TH, Seo GS, Kim YC, Sohn DH (2003)
The chalcone butein from Rhus verniciflua shows antifibrogenic activity. Planta Med 69:990–
994
37. Lee SH, Seo GS, Sohn DH (2004) Inhibition of lipopolysaccharide-induced expression of
inducible nitric oxide synthase by butein in RAW 264.7 cells. Biochem Biophys Res Commun
323(1):125–132
38. Lee SH, Seo GS, Jin XY, Ko G, Sohn DH (2007) Butein blocks tumor necrosis factor
alpha-induced interleukin 8 and matrix metalloproteinase 7 production by inhibiting p38
kinase and osteopontin mediated signaling events in HT-29 cells. Life Sci 81(21–22):1535–
1543
39. Lee SH, Choi SC, Kim KS, Park JW, Lee SH, Yoon SW (2010) Shrinkage of gastric cancer in
an elderly patient who received Rhus verniciflua Stokes extract. J Altern Complement Med 16
(4):497–500
40. Lee DS, Li B, Im NK, Kim YC, Jeong GS (2013) 4,2’,5’-trihydroxy-4’-methoxychalcone
from Dalbergia odorifera exhibits anti-inflammatory properties by inducing heme
oxygenase-1 in murine macrophages. Int Immunopharmacol 16(1):114–121
41. Lee SD, Choe JW, Lee BJ, Kang MH, Joo MK, Kim JH, Yeon JE, Park JJ, Kim JS, Bak YT
(2015) Butein effects in colitis and interleukin-6/signal transducer and activator of
transcription 3 expression. World J Gastroenterol 21(2):465–474
42. Li Y, Ma C, Qian M, Wen Z, Jing H, Qian D (2014) Butein induces cell apoptosis and
inhibition of cyclooxygenase2 expression in A549 lung cancer cells. Mol Med Rep 9(2):763–
767
43. Liao XL, Luo JG, Kong LY (2013) Flavonoids from Millettia nitida var. hirsutissima with
their anticoagulative activities and inhibitory effects on NO production. J Nat Med 67(4):856–
861
44. Lim SS, Jung SH, Ji J, Shin KH, Keum SR (2001) Synthesis of avonoids and their effects on
aldose reductase and sorbitol accumulation in streptozotocin-induced diabetic rat tissues.
J Pharm Pharmacol 53:653–668
45. Lin CN, Lin TH, Hsu MF, Wang JP, Ko FN, Teng CM (1997) 2’,5’-Dihydroxychalcone as a
potent chemical cyclooxygenase inhibitor mediator and cyclooxygenase inhibitor. J Pharm
Pharmacol 49:530–536
46. Liu SC, Chen C, Chung CH, Wang PC, Wu NL, Cheng JK, Lai YW, Sun HL, Peng CY,
Tang CH, Wang SW (2014) Inhibitory effects of butein on cancer metastasis and bioenergetic
modulation. J Agric Food Chem 62(37):9109–9117
47. Lu M, Wang S, Han X, Lv D (2013) Butein inhibits NF-kappaB activation and reduces
infiltration of inflammatory cells and apoptosis after spinal cord injury in rats. Neurosci Lett
542:87–91
48. Martineau LC (2012) Large enhancement of skeletal muscle cell glucose uptake and
suppression of hepatocyte glucose-6-phosphatase activity by weak uncouplers of oxidative
phosphorylation. Biochim Biophys Acta 1820(2):133–150
49. Mendis S, Puska P, Norrving B (2011) Global Atlas on cardiovascular disease prevention and
control. World Health Organization, 3–18
432 Z. Song et al.
50. Moon DO, Choi YH, Moon SK, Kim WJ, Kim GY (2010) Butein suppresses the expression of
nuclear factor-kappa B-mediated matrix metalloproteinase-9 and vascular endothelial growth
factor in prostate cancer cells. Toxicol In Vitro 24(7):1927–1934
51. National Center for Biotechnology Information (2005) CID=5281222.PubChem compound
database. https://pubchem.ncbi.nlm.nih.gov/compound/5281222
52. Neergheen VS, Bahorun T, Taylor EW, Jen LS, Aruoma OI (2010) Targeting specific cell
signaling transduction pathways by dietary and medicinal phytochemicals in cancer
chemoprevention. Toxicology 278(2):229–241
53. Nerya O, Musa R, Khatib S, Tamir S, Vaya J (2004) Chalcones as potent tyrosinase inhibitors:
the effect of hydroxyl positions and numbers. Phytochemistry 65(10):1389–1395
54. Orlikova B, Schnekenburger M, Zloh M, Golais F, Diederich M, Tasdemir D (2012) Natural
chalcones as dual inhibitors of HDACs and NF-kappaB. Oncol Rep 28(3):797–805
55. Pandey MK, Sandur SK, Sung B, Sethi G, Kunnumakkara AB, Aggarwal BB (2007) Butein, a
tetrahydroxychalcone, inhibits nuclear factor (NF)-kappaB and NF-kappaB-regulated gene
expression through direct inhibition of IkappaBalpha kinase beta on cysteine 179 residue.
J Biol Chem 282(24):17340–17350
56. Paterson I, Anderson E (2005) The renaissance of natural products as drug candidates. Science
310:451–453
57. Rajendran P, Ong TH, Chen L, Li F, Shanmugam MK, Vali S, Abbasi T, Kapoor S,
Sharma A, Kumar AP, Hui KM, Sethi G (2011) Suppression of signal transducer and activator
of transcription 3 activation by butein inhibits growth of human hepatocellular carcinoma
in vivo. Clin Cancer Res 17(6):1425–1439
58. Rasheed Z, Akhtar N, Khan A, Khan KA, Haqqi TM (2010) Butrin, isobutrin, and butein from
medicinal plant Butea monosperma selectively inhibit nuclear factor-kappaB in activated
human mast cells: suppression of tumor necrosis factor-alpha, interleukin (IL)-6, and IL-8.
J Pharmacol Exp Ther 333(2):354–363
59. Selvam C, Jachak SM, Bhutani KK (2004) Cyclooxygenase inhibitory flavonoids from the
stem bark of Semecarpus anacardium Linn. Phytother Res 18:582–584
60. Semwal RB, Semwal DK, Combrinck S, Viljoen A (2015) Butein: from ancient traditional
remedy to modern nutraceutical. Phytochem Lett 11:188–201
61. Song DG, Lee JY, Lee EH, Jung SH, Nho CW, Cha KH, Koo SY, Pan CH (2010) Inhibitory
effects of polyphenols isolated from Rhus verniciflua on Aldo-keto reductase family 1 B10.
BMB Rep 43:268–272
62. Song NJ, Yoon HJ, Kim KH, Jung SR, Jang WS, Seo CR, Lee YM, Kweon DH, Hong JW,
Lee JS, Park KM, Lee KR, Park KW (2013) Butein is a novel anti-adipogenic compound.
J Lipid Res 54(5):1385–1396
63. Sung J, Lee J (2015) Anti-inflammatory activity of butein and luteolin through suppression of
NFkappaB activation and induction of heme oxygenase-1. J Med Food 18(5):557–564
64. Szuster-Ciesielska A, Mizerska-Dudka M, Daniluk J, Kandefer-Szerszen M (2013) Butein
inhibits ethanol-induced activation of liver stellate cells through TGF-beta, NFkappaB, p38,
and JNK signaling pathways and inhibition of oxidative stress. J Gastroenterol 48(2):222–237
65. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J (2007) Free radicals and
antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 39
(1):44–84
66. Vitaglione P, Morisco F, Caporaso N, Fogliano V (2005) Dietary antioxidant compounds and
liver health. Crit Rev Food Sci Nutr 44(7–8):575–586
67. Wang Y, Chan FL, Chen S, Leung LK (2005) The plant polyphenol butein inhibits
testosterone-induced proliferation in breast cancer cells expressing aromatase. Life Sci 77
(1):39–51
68. Wang Z, Lee Y, Eun JS, Bae EJ (2014) Inhibition of adipocyte inflammation and macrophage
chemotaxis by butein. Eur J Pharmacol 738:40–48
69. Woo SW, Lee SH, Kang HC, Park EJ, Zhao YZ, Kim YC, Sohn DH (2003) Butein suppresses
myofibroblastic differentiation of rat hepatic stellate cells in primary culture. J Pharm
Pharmacol 55(3):347–352
17 Butein and Its Role in Chronic Diseases 433
70. Xu HX, Wan M, Dong H, But PP, Foo LY (2000) Inhibitory activity of flavonoids and tannins
against HIV-1 protease I. Biol Pharm Bull 23(9):1072–1076
71. Yadav VR, Prasad S, Sung B, Aggarwal BB (2011) The role of chalcones in suppression of
NF-kappaB-mediated inflammation and cancer. Int Immunopharmacol 11(3):295–309
72. Yang LH, Ho YJ, Lin JF, Yeh CW, Kao SH, Hsu LS (2012) Butein inhibits the proliferation of
breast cancer cells through generation of reactive oxygen species and modulation of ERK and
p38 activities. Mol Med Rep 6(5):1126–1132
73. Yang PY, Hu DN, Lin IC, Liu FS (2015) Butein shows cytotoxic effects and induces apoptosis
in human ovarian cancer cells. Am J Chin Med 43(4):769–782
74. Yit CC, Das NP (1994) Cytotoxic effect of butein on human colon adenocarcinoma cell
proliferation. Cancer Lett 82:65–72
75. Yu SM, Cheng ZJ, Kuo SC (1995) Endothelium-dependent relaxation of rat aorta by butein, a
novel cyclic AMP-specific phosphodiesterase inhibitor. Eur J Pharmacol 280:69–77
76. Yu H, Pardoll D, Jove R (2009) STATs in cancer inflammation and immunity: a leading role
for STAT3. Nat Rev Cancer 9(11):798–809
77. Zhang K, Wong KP (1997) Glutathione conjugation of chlorambucil: measurement and
modulation by plant polyphenols. Biochem J 325:417–422
78. Zhang L, Chen W, Li X (2008) A novel anticancer effect of butein: inhibition of invasion
through the ERK1/2 and NF-kappa B signaling pathways in bladder cancer cells. FEBS Lett
582(13):1821–1828
Chapter 18
Garcinol and Its Role in Chronic Diseases
Abstract The various bioactive compounds isolated from leaves and fruits of
Garcinia sps plants, have beencharacterized and experimentally demonstrated to be
anti-oxidant, anti-inflammatory and anti-cancer in nature.Garcinol, a polyiso-
prenylated benzophenone, obtained from plant Garcinia indica has been found to be
aneffective inhibitor of several key regulatory pathways (e.g., NF-kB, STAT3 etc.)
in cancer cells, thereby being ableto control malignant growth of solid tumours
in vivo. Despite its high potential as an anti-neoplastic modulator ofseveral cancer
types such as head and neck cancer, breast cancer, hepatocellular carcinoma,
prostate cancer,colon cancer etc., it is still in preclinical stage due to lack of sys-
tematic and conclusive evaluation ofpharmacological parameters. While it is
promising anti-cancer effects are being positively ascertained fortherapeutic
development, studies on its effectiveness in ameliorating other chronic diseases
such ascardiovascular diseases, diabetes, allergy, neurodegenerative diseases etc.,
though seem favourable, are veryrecent and require in depth scientific investigation.
18.1 Introduction
Fig. 18.1 a Image of Garcinia indica tree. b Ripe fruits of Garcinia indica. c Chemical structure
of garcinol
practices, leaves and fruits of this plant have been used for treatment of inflam-
matory ailments and digestive disorders [1]. As per Ayurvedic classical texts, it has
been mentioned as cardiac tonic, anti-helminthic and used in intestinal disorders,
hemorrhoids, tumors, etc. [2]. Many groups have put efforts to identify and char-
acterize the biological function and medicinal value of different phytochemicals
obtained from this plant. Different studies have independently demonstrated that
garcinol possesses antibacterial, antioxidant, gastroprotective, and most impor-
tantly, antineoplastic properties. Recent investigations have revealed that garcinol
treatment significantly affects multiple biological pathways, suggesting its potent
therapeutic role in multitude of human disorders.
The diseases which are persistent and long lasting, such as cancer, cardiovascular
diseases, diabetes, arthritis, neurodegenerative diseases, allergies, etc., are included in
the group of chronic diseases. With current advancement in medical science, complete
cure is not possible for the pathological conditions seen in chronic diseases. However,
with medical intervention controlling measures could be taken so as to alleviate the
pathological severity or retard the progression of the disease. As a result the inevitable
outcome of mortality in case of such diseases could be delayed. Several of the chronic
diseases could be prevented on the grounds of healthy lifestyle. Thus, self-awareness
and personal management of illness also could help to avoid complications and
increase survival. Nevertheless, researchers have been putting constant efforts to find
cures for such diseases. In the field of drug development for human diseases, naturally
available compounds and small molecules derived from medicinal plant sources have
been under investigation in last few decades. On a similar basis, there have been
several preclinical studies carried out with garcinol testifying and underscoring its
value with therapeutic promises in manifold human disorders including chronic
diseases, which will be discussed in subsequent sections.
regulating its anticancer activity in context of oral cancer. In this study it has been
revealed that 13,14-dimethoxy derivative of garcinol has very little effect in regulating
cell cycle and apoptosis of SCC15 cell line compared to unmethylated garcinol. The
mechanism behind this difference has been attributed to the ability of garcinol to bind
and inhibit 5-lipoxygenase (5-Lox), where hydroxyl groups at C13 and C14 seem to
be critical in interaction with the catalytic domain of 5-Lox [3]. Importance of C-13
and C-14 has been further shown by another study, where substitution of C-13 and
C-14 of isogarcinol (an intramolecular cyclization product of garcinol) could enhance
the specificity of the molecule in context of inhibition of histone acetyltransferase
(HAT) activity [4]. It has been further discussed in Sect. 18.7.
Pharmacokinetic Properties: According to the calculation by Guide to
Pharmacology (IUPHAR/BPS) online tool, Garcinol has four hydrogen bond
acceptors and three hydrogen bond donors. It has 10 rotatable bonds. Its 2-D
structure would have topological polar surface area of 111.9 Å2. It displays an
octanol–water partition coefficient: log P of 10.07. The number of Lipinski’s rules
broken is 2, making it pharmacologically less amenable to be an orally active drug,
which is a theoretical prediction.
There have been very few studies toward understanding of bioavailability of gar-
cinol. According to one investigation, effect of garcinol seems to be more effective
in the absence of serum in in vitro cell culture system. The addition of 10 % FBS
(fetal bovine serum) to medium leads to approximately 10-fold decrease in IC50
value of garcinol for HCT116 cell growth. Addition of BSA (bovine serum albu-
min) protein to media showed similar effect, indicating interaction of garcinol with
serum proteins. However, no such effect was observed with cambogin (found in
Garcinia cambogia, having similar but not identical structure with that of garcinol).
Cellular uptake assessment studies showed that intracellular garcinol level in HT-29
and HCT116 cells was 2–5-fold more than that of cambogin after incubation in
serum-free Hank’s balanced salt solution for 1 h. However, in the presence of FBS
the intracellular level of garcinol was dramatically reduced under similar experi-
mental conditions [5].
Pharmacokinetic properties of garcinol with respect to its absorption, metabo-
lism, tissue distribution, excretion and physiological toxicity, etc. are needed to be
investigated in animal model before considering for clinical trial toward therapeutic
advancement, despite its promising anticancer properties.
Garcinol has been demonstrated to inhibit HATs such as p300 and PCAF both
in vitro and in vivo; as a result it could suppress HAT-dependent transcription from
chromatin. It was further shown by microarray analysis that inhibition of HAT with
treatment of garcinol could suppress expression of majority of genes tested at global
level in HeLa cells. Several of downregulated genes were revealed to be
438 A.K. Behera et al.
NF-kB
STAT3
Wnt-Β-catenin Proliferative
Gli1 pathway
Cyclin D1
Notch 1
MET E-cadherin FAK
Epigenetic
p300/CBP
pathway
DR4, DR5
Vimentin
Cytosolic
Apoptotic
Garcinol ZEB 1 EMT
cytochrome c
pathway ZEB 2
Bax, tBid
Cleaved caspase 3 Bcl2 Anti-apoptotic
XIAP pathway
FLIP
MMP7
Metastatic
VEGF
pathway
NF-kB
iNOS Inflammatory
NO pathway
COX2
Fig. 18.2 Different cellular pathways positively and negatively regulated by garcinol in cell. DR4
death receptor 4, Bax Bcl2-associated X Protein, tBid truncated BH3 interacting-domain death
agonist, NF-kB nuclear factor kappa light chain enhancer of activated B cells, STAT3 signal
transducer and activator of transcription 3, FAK focal adhesion kinase, CBP CREB-binding
protein, ZEB1 zinc finger E-box-binding homeobox 1, Bcl2 B-cell lymphoma 2, XIAP X-linked
inhibitor of apoptosis protein, FLIP FLICE-like inhibitory protein, MMP7 matrix
metalloproteinase-7, VEGF vascular endothelial growth factor, iNOS inducible nitric oxide
synthase, NO nitric oxide, COX-2 cyclooxygenase-2
18 Garcinol and Its Role in Chronic Diseases 439
Nuclear localization,
Phosphorylation Phosphorylation
IKK IkBα NF-kB
Gene regulation
JAK2 p300
Phosphorylation
Acetylation
Garcinol
STAT3 STAT3
STAT3 STAT3
STAT3 STAT3
Fig. 18.3 Scheme to show the mechanism through which garcinol suppresses NF-kB and STAT3
pathways. Garcinol inhibits IKK activity, resulting in reduced degradation and increased
accumulation of IkBa, which in turn phosphorylates NF-kB and leads to degradation of NF-kB.
Garcinol inhibits both phosphorylation and acetylation of STAT3 by JAK2 and p300, respectively,
which reduces the nuclear localization of STAT3. Garcinol also directly binds to STAT3 and
impairs the dimerization and thus DNA-binding and transcriptional regulation ability of STAT3
440 A.K. Behera et al.
Hep3B cells (deficient for p53), where it has been demonstrated to activate death
receptor and mitochondrial apoptotic pathway with significant activation of caspase
9 and 3. It could lead to accumulation of reactive oxygen species (ROS), endo-
plasmic reticulum (ER) stress modulator GADD153 (growth arrest and DNA
damage inducible gene 153), increased Bax2/Bcl-2 ratio, elevated tBid (truncated
Bid), and caspase 8 in cancer cell. This study highlights the therapeutic application
of garcinol in the context of p53-independent apoptosis [21]. Garcinol also sup-
pressed the growth of prostate cancer cell lines such as LNCaP, C4-2B, and PC3
in vitro and could induce apoptotic cell death. This chemopreventive nature of
garcinol was demonstrated to occur via inhibition of NF-kB pathway [13]. Study
with colorectal cancer cell line HT-29 shows that garcinol can lead to inhibition of
tyrosine phosphorylation of focal adhesion kinase (FAK), which is a mediator of
integrin-regulated intracellular signaling in adherent cells, thus impairing cellular
proliferation and migration. It also inhibited expression of MMP7 (matrix metal-
loproteinase 7) in HT-29 cells, thereby leading to inhibition of invasiveness of the
cancerous cells. Researchers were also able to demonstrate that it could lead to
inhibition of growth promoting c-Src, MAPK/ERK, and PI3K/Akt signaling
pathways and can cause release of cytochrome c from mitochondria to cytosol and
induction of apoptosis in HT-29 colorectal cells [22].
Diabetes: Diabetes is a pathological condition characterized by prolonged and
high levels of sugar in blood of the patient. Either inadequate production of insulin
or inability of cells of body to respond to insulin for proper metabolism of sugar or
both could lead to diabetes. Diabetic condition is known to cause damage in small
blood vessels to the eye, kidney, and nerves causing diabetic retinopathy, diabetic
nephropathy, and diabetic neuropathy, respectively, with damage to respective
organs. Investigations have revealed marked hyperacetylation of histones in several
organs of diabetic rats such as retina. One study has successfully targeted histone
acetylation in retinal Müller glia cells grown in a diabetes-like concentration of
glucose, which would mimic pathogenesis of diabetic retinopathy. In this report the
researchers show that garcinol treatment could lead to reduction in histone acety-
lation, consequently suppressing expression of the pro-inflammatory factors
implicated in diabetic retinopathy [23]. With a similar perspective, curcumin analog
C66 has been found to prevent diabetic nephropathy in mice via inhibition of HAT
activity of p300/CBP, which seems to be one of important mechanisms modulated
by curcumin and its analogs to exert antidiabetic effects [24–26], further empha-
sizing the significance of garcinol in the prospects of treatment of diabetic com-
plications as a modulator of HATs. Another line of research indicates that garcinol
could also target the process of adipogenesis and reduce accumulation of lipids in
3T3-L1 cells [27]. With further research anti-adipogenic effect of garcinol could be
exploited to manage obesity, which is known to increase the risk of type 2 diabetes.
Cardiovascular Diseases: Many cardiovascular diseases result in cardiac fibrosis
which in turn leads to thickening and stiffening of cardiac muscle in the progression
to heart failure. It has been found that excessive expression of collagen-1 (which
normally provides structural support to heart) in cardiac fibroblasts in fibrotic
442 A.K. Behera et al.
Garcinol is still in preclinical stage. There have been numerous in vitro studies with
cell lines of human origin as well as in animal models for different human diseases,
to assess efficacy of garcinol toward development of chemoprevention modalities.
However, there has not been any systematic study on human as a whole organism.
A commercially available medicinal product called GarCitrin® (an extract obtained
from Garcinia cambogia) contains 5 % of garcinol as constituent along with
minimum of 50 % of hydroxycitric acid (HCA). The major purpose of using
GarCitrin® relies on activity of HCA to block biosynthesis of fatty acids and
lipogenesis thus aiding in weight management in an individual [37]. Although it is
444 A.K. Behera et al.
High cytotoxicity of garcinol has prompted several groups to explore the avail-
ability of bioactive compounds with analogous chemical structures as well as
synthesizing derivatives on garcinol scaffold with better biological utility.
According to one such study, mono-substituted isogarcinol (IG: an intramolecular
cyclization product of garcinol) derivatives, LTK-13 (14-isopropoxy IG), LTK-14
(14-methoxy IG), and LTK-19 (13,14 disulfoxy IG) were specific inhibitors of
p300-HAT compared to parental compound, which inhibits both p300 and
PCAF HAT activity. Further investigation with the derivative LTK14, which is
specific inhibitor of p300 and non-toxic to cells, showed that it could suppress HIV
multiplication in T lymphocytes via HAT inhibition [4]. Another study, in context
of cancer, showed that two oxidative derivatives of garcinol namely, garcim-1 and
garcim-2, possessed greater inhibitory potential on growth of colon cancer cells [5].
Compounds from different species of Garcinia genus having similar structures
also have been isolated and found to be exhibiting similar biological activities,
18 Garcinol and Its Role in Chronic Diseases 445
Name of Garcinia sp. Name of the Chemical structure Biological activity References
compound isolated
Garcinia indica, Garcinia Isogarcinol (a) Anticancer, pro-apoptotic (a) Pieme et al.
ovalifolia, Garcinia (b) Immuno-suppressant [40]
mangostana OH (b) Fu et al. [41]
HO O O H
H
O O
H
O OH
(continued)
Table 18.3 (continued)
18
Name of Garcinia sp. Name of the Chemical structure Biological activity References
compound isolated
Garcinia xanthochymus Xanthochymol Anticancer Matsumoto et al.
Pro-apoptotic [49],
OH Einbond et al.
HO O O H
[50]
H
O OH
O O H
Garcinol and Its Role in Chronic Diseases
O OH
O OH
Garcinia hanburyi Gambogic acida Anticancer and pro-apoptotic (a) Wang et al.
effect on [53]
OH (a) Lung cancer (b) Shi et al. [54]
O
O O O O (b) Chronic myelogenous (c) Wen et al. [55]
leukemia
(c) Colorectal cancer
447
OH O
(continued)
Table 18.3 (continued)
448
Name of Garcinia sp. Name of the Chemical structure Biological activity References
compound isolated
Garcinia kola Kolaviron OH O Antidiabetic and hypoglycemic Iwu et al. [56],
OH Oyenihi et al. [57]
OH
HO O
H
O
O
HO O
OH
OH
Garcinia mangostana Alpha-Mangostin (a) Anti-inflammatory, (a) Janhom and
cytoprotective and anti-apoptotic Dharmasaroja
OH O
in SH-SY5Y cells [58]
O
(b) Anticancer (b) Hafeez et al.
HO O OH
(c) Anti-invasive [59]
(c) Wang et al.
[60]
Garcinia nujiangensis Nujiangexanthone Antiasthmatic effect Lu et al. [61]
A OH O
HO
O O OH
OH
a
Approved by the Chinese Food and Drug Administration for the treatment of lung cancer and is in Phase II clinical trials [62]
A.K. Behera et al.
18 Garcinol and Its Role in Chronic Diseases 449
18.8 Conclusions
Among several natural compounds isolated from fruit rind of Garcinia indica,
garcinol has been found to be the major component responsible for medicinal utility
of the plant. Initial studies showed potent antioxidant and anti-inflammatory
properties of garcinol. Further, in vitro and in vivo studies in animal models have
demonstrated its role as an effective anticancer agent in multitude of cancers, such
as prostate, colon, head and neck, pancreatic, and breast cancers. Therapeutic
potential of garcinol has been found not to be limited to cancers alone. Several
scientific researches indicate that it could exhibit benefits in many other chronic
diseases such as cardiovascular disorders, diabetes, neurodegenerative diseases, and
certain allergies such as asthma. However, majority of investigations carried out
with garcinol have explored and emphasized on its promising antineoplastic
properties. Researchers have identified and elucidated the molecular targets of
garcinol in different cancer cells and explained the basis of antitumor activity of
garcinol. It could suppress the growth in cancer cells via inhibition of several of
pro-survival pathways such as NF-kB, STAT3, and Akt/PI3K pathways and
simultaneously can induce apoptotic cell death. Additionally, it inhibits epithelial to
mesenchymal transition and metastasis of cancer cells and favors mesenchymal to
epithelial transition. Despite several animal studies justifying therapeutic role of
garcinol in last decade, it is still in preclinical stage of drug development due to lack
of proper pharmacokinetic studies. Further investigations are required to determine
toxicological aspects of garcinol and assess the parameters encompassing its
bio-metabolism such as absorption, systemic and/or localized distribution, and
excretion at the level of whole organism. It is important to understand the bio-
logically effective dosage or concentrations of garcinol for a specific disease, route
of administration, and temporal scheme of administration for better bioavailability
in whole organism, for ultimate development toward clinical applications. Proper
pharmacological assessment of garcinol could shed light into its relative propensity
of being effective as a future drug and encourage researchers to look for structural
analogs and synthesize derivatives of garcinol to improve on the limitations and
make it more effective and amenable for clinical therapy.
References
1. Baliga MS, Bhat HP, Pai RJ et al (2011) The chemistry and medicinal uses of the underutilized
Indian fruit tree Garcinia indica Choisy (kokum): a review. Food Res Intern 44(7):1790–1799
2. Charaka samhita text, suthrasthaana, chapter 4th and 27th, Bhava prakasha text
3. Han CM, Zhou XY, Cao J et al (2015) 13,14-Dihydroxy groups are critical for the anti-cancer
effects of garcinol. Bioorg Chem 60:123–129
4. Mantelingu K, Reddy BA, Swaminathan V et al Specific inhibition of p300-HAT alters global
gene expression and represses HIV replication. Chem Biol 14(6):645–57
5. Hong J, Kwon SJ, Sang S et al (2007) Effects of garcinol and its derivatives on intestinal cell
growth: inhibitory effects and autoxidation-dependent growth-stimulatory effects. Free Radic
Biol Med 42(8):1211–1221
6. Balasubramanyam K, Altaf M, Varier RA et al (2004) Polyisoprenylated benzophenone,
garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and
alters global gene expression. J Biol Chem 279(32):33716–33726
7. Huang CC, Lin CM, Huang YJ et al (2015) Garcinol downregulates Notch1 signaling via
modulating miR-200c and suppresses oncogenic properties of PANC-1 cancer stem-like cells.
Biotechnol Appl Biochem. doi:10.1002/bab.1446
8. Tsai ML, Chiou YS, Chiou LY et al (2014) Garcinol suppresses inflammation-associated
colon carcinogenesis in mice. Mol Nutr Food Res 58(9):1820–1829
9. Yoshida K, Tanaka T, Hirose Y et al (2005) Dietary garcinol inhibits 4-nitroquinoline
1-oxide-induced tonguecarcinogenesis in rats. Cancer Lett 221(1):29–39
10. Ahmad A, Sarkar SH, Aboukameel A et al (2012) Anti-cancer action of garcinol in vitro and
in vivo is in part mediated through inhibition of STAT-3 signaling. Carcinogenesis 33
(12):2450–2456
11. Sethi G, Chatterjee S, Rajendran P et al (2014) Inhibition of STAT3 dimerization and
acetylation by garcinol suppresses the growth of human hepatocellular carcinoma in vitro and
in vivo. Mol Cancer 13:66
12. Ahmad A, Sarkar SH, Bitar B et al (2012) Garcinol regulates EMT and Wnt signaling
pathways in vitro and in vivo, leading to anticancer activity against breast cancer cells. Mol
Cancer Ther 11(10):2193–2201
13. Ahmad A, Wang Z, Wojewoda C et al (2011) Garcinol-induced apoptosis in prostate and
pancreatic cancer cells is mediated by NF- kappaB signaling. Front Biosci (Elite Ed) 3:1483–
1492
14. Ahmad A, Wang Z, Ali R et al (2010) Apoptosis-inducing effect of garcinol is mediated by
NF-kappaB signaling in breast cancer cells. J Cell Biochem 109(6):1134–1141
15. Pan MH, Chang WL, Lin-Shiau SY et al (2001) Induction of apoptosis by garcinol and
curcumin through cytochrome c release and activation of caspases in human leukemia HL-60
cells. J Agric Food Chem 49(3):1464–1474
16. Prasad S, Ravindran J, Sung B et al (2010) Garcinol potentiates TRAIL-induced apoptosis
through modulation of death receptors and antiapoptotic proteins. Mol Cancer Ther 9(4):856–
868
17. Oike T, Ogiwara H, Torikai K et al (2012) Garcinol, a histone acetyltransferase inhibitor,
radiosensitizes cancer cells by inhibiting non-homologous end joining. Int J Radiat Oncol Biol
Phys 84(3):815–821
18. Chen X, Zhang X, Lu Y et al (2012) Chemoprevention of 7,12-dimethylbenz[a]anthracene
(DMBA)-induced hamster cheek pouch carcinogenesis by a 5-lipoxygenase inhibitor,
garcinol. Nutr Cancer 64(8):1211–1218
19. Li F, Shanmugam MK, Chen L (2013) Garcinol, a polyisoprenylated benzophenone modulates
multiple proinflammatory signaling cascades leading to the suppression of growth and survival
of head and neck carcinoma. Cancer Prev Res (Phila) 6(8):843–54
20. Parasramka MA, Gupta SV (2012) Synergistic effect of garcinol and curcumin on
antiproliferative and apoptotic activity in pancreatic cancer cells. J Oncol 2012:709739
18 Garcinol and Its Role in Chronic Diseases 451
21. Cheng AC, Tsai ML, Liu CM et al (2010) Garcinol inhibits cell growth in hepatocellular
carcinoma Hep3B cells through induction of ROS-dependent apoptosis. Food Funct 1(3):301–
307
22. Liao CH, Sang S, Ho CT et al (2005) Garcinol modulates tyrosine phosphorylation of FAK
and subsequently induces apoptosis through down-regulation of Src, ERK, and Akt survival
signaling in human colon cancer cells. Cell Biochem 96(1):155–169
23. Kadiyala CS, Zheng L, Du Y et al (2012) Acetylation of retinal histones in diabetes increases
inflammatory proteins: effects of minocycline and manipulation of histone acetyltransferase
(HAT) and histone deacetylase (HDAC). J Biol Chem 287(31):25869–25880
24. Wang Y, Wang Y, Luo M et al (2015) Novel curcumin analog C66 prevents diabetic
nephropathy via JNK pathway with the involvement of p300/CBP-mediated histone
acetylation. Biochim Biophys Acta 1852(1):34–46
25. Yun JM, Jialal I, Devaraj S (2011) Epigenetic regulation of high glucose-induced
proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem
22(5):450–458
26. Pham TX, Lee J (2012) Dietary regulation of histone acetylases and deacetylases for the
prevention of metabolic diseases. Nutrients 4(12):1868–1886
27. Hsu CL, Lin YJ, Ho CT et al (2012) Inhibitory effects of garcinol and pterostilbene on cell
proliferation and adipogenesis in 3T3-L1 cells. Food Funct 3(1):49–57
28. Chan EC, Dusting GJ, Guo N et al (2010) Prostacyclin receptor suppresses cardiac fibrosis:
role of CREB phosphorylation. J Mol Cell Cardiol 49(2):176–185
29. Balasubramanyam K, Swaminathan V, Ranganathan A et al (2003) Small molecule
modulators of histone acetyltransferase p300. J Biol Chem 278(21):19134–19140
30. Peng C, Zhang W, Zhao W, Zhu J, Huang X, Tian J (2015) Alcohol-induced histone H3K9
hyperacetylation and cardiac hypertrophy are reversed by a histone acetylases inhibitor
anacardic acid in developing murine hearts. Biochimie 113:1–9
31. Davidson SM, Townsend PA, Carroll C et al (2005) The transcriptional coactivator p300 plays
a critical role in the hypertrophic and protective pathways induced by phenylephrine in cardiac
cells but is specific to the hypertrophic effect of urocortin. ChemBioChem 6(1):162–170
32. Weng MS, Liao CH, Yu SY et al (2011) Garcinol promotes neurogenesis in rat cortical
progenitor cells through the duration of extracellular signal-regulated kinase signaling. J Agric
Food Chem 59(3):1031–1040
33. Liao CH, Ho CT, Lin JK (2005) Effects of garcinol on free radical generation and NO
production in embryonic rat cortical neurons and astrocytes. Biochem Biophys Res Commun
329(4):1306–1314
34. Cui ZL, Gu W, Ding T et al (2013) Histone modifications of Notch1 promoter affect lung
CD4+ T cell differentiation in asthmatic rats. Int J Immunopathol Pharmacol 26(2):371–381
35. Tanaka T, Kohno H, Shimada R et al (2000) Prevention of colonic aberrant crypt foci by
dietary feeding of garcinol in male F344 rats. Carcinogenesis 21(6):1183–1189
36. Wang Y, Tsai ML, Chiou LY et al (2015) Antitumor activity of garcinol in human prostate
cancer cells and xenograft mice. J Agric Food Chem 63(41):9047–9052
37. Heymsfield SB, Allison DB, Vasselli JR et al (1998) Garcinia cambogia (hydroxycitric acid)
as a potential antiobesity agent: a randomized controlled trial. JAMA 280(18):1596–1600
38. Padhye S, Ahmad A, Oswal N et al (2009) Emerging role of Garcinol, the antioxidant
chalcone from Garcinia indica Choisy and its synthetic analogs. J Hematol Oncol 2:38
39. Li F, Shanmugam MK, Siveen KS et al (2015) Garcinol sensitizes human head and neck
carcinoma to cisplatin in a xenograft mouse model despite downregulation of proliferative
biomarkers. Oncotarget 6(7):5147–5163
40. Pieme CA, Ambassa P, Yankep E et al (2015) Epigarcinol and isogarcinol isolated from the
root of Garcinia ovalifolia induce apoptosis of human promyelocytic leukemia (HL-60 cells).
BMC Res Notes 8(1):700
41. Fu Y, Zhou H, Wang M et al (2014) Immune regulation and anti-inflammatory effects of
isogarcinol extracted from Garcinia mangostana L. against collagen-induced arthritis. J Agric
Food Chem 62(18):4127–4134
452 A.K. Behera et al.
Keywords Morin Chronic diseases Anti-inflammatory Antioxidant Cellular
signaling
19.1 Introduction
Affordable cost, presence mostly in the consumables, and minimal side effects make
the naturally occurring compounds interesting and attractive for pharmacological
study in recent years. Plants produce diverse types of low molecular weight products
mainly for the defense purpose. Among them, the group of secondary metabolites
related to a polyphenolic group has been named flavonoids and are of great interest
due to their incredible pharmacological properties [14, 24]. These compounds are
consumed regularly as a part of the diet, present in flowers, tea, red wine, fruits, nuts,
herbs, vegetables, seeds, spices, stems, etc. and are known to possess a number of
biological and pharmacological activities like anti-hepatotoxic, anti-inflammatory,
antiulcer, antioxidant, etc., and also have the ability to inhibit various enzyme
activities [12, 27]. Among these, morin [morin hydrate:2-(2,4-dihydroxyphenyl)-
3,5,7-trihydroxy-4H-1-benzopy ran-4-one; 3,5,7,20,40 pentahydroxyflavone]
(Fig. 19.1) is belonging to the group of flavonols (a class of flavonoids having
3-hydroxyflavon backbone) found in the branches of white mulberry (Morus alba L),
osage orange (Maclura pomifera), almond (Psidium guajava), fig (Chlorophora
tinctoria), mill (Prunus dulcis), old fustic (Maclura tinctoria), and other family
members of Moraceae along with sweet chestnut (Castanea sativa, family Fagaceae)
[4, 35]. This compound exhibits different types of pharmacological activities, like free
radical scavenging, anti-inflammatory, xanthine oxidase inhibitor property, protec-
tive effect on DNA from damage caused by free radicals, prevention of low-density
lipoprotein oxidation, anticancer activity, etc. [14, 35]. Further studies both in in vitro
and in vivo indicate that it possesses numerous add on health benefits. However, no
article has yet been published that comprises and explains its botanical origin and
pharmacological activities in spite of the current research progress made on
Morin has a molecular weight of 302.2357 g/mol, exact mass of 302.042653 g/mol,
monoisotopic mass of 302.042653 g/mol, molecular formula of C15H10O7, XLogP3
of 1.5, hydrogen bond donor count of 5, hydrogen bond acceptor count of 7,
rotatable bond count of 1, topological polar surface area of 127 A2, heavy atom
456 K. Sinha et al.
count of 22, formal charge of 0, complexity of 488, isotope atom count of 0, defined
atom stereocenter count of 0, undefined atom stereocenter count of 0, defined bond
stereo center count of 0, undefined bond stereo center count of 0, a covalently
bonded unit count of 1, and experimental melting point 303.5 °C [5]. Morin is a
naturally occurring molecule (can also be synthesized) and it inhibits or retard the
oxidation of a substance to which it is added. It counteracts the harmful and
damaging effects of oxidation in animal tissues. The structure [Fig. 19.1] of morin
hydrate shows that it is an isomeric form of quercetin. Quercetin and morin both
have OH in position 3, a carbonyl group in position 4, and a resorcinol moiety.
However, there is a difference in the hydroxylation pattern on B-ring. In morin, the
hydroxylation is at the meta-position (in Morin), whereas it is in the ortho position
in quercetin. For all its groups and an ortho hydroxylation pattern on B-ring,
quercetin is regarded to have the highest antioxidant potential of the flavonoids but
morin hydrate has also been demonstrated to have higher effectiveness of certain
oxidative processes. Morin is soluble in methanol (50 mg ml−1) and generously
soluble in alcohol whereas faintly soluble in ether and acetic acid. It is also soluble
in aqueous alkaline solutions and gives intense yellow color. This color changes to
brown upon water exposure. It is also soluble in water (0.25 mg/ml, 20 °C;
0.94 mg/ml, 100 °C). In respect to pharmacokinetic study, Hou et al. investigated
and compared the pharmacokinetics of morin and its isomer quercetin in rats. Parent
forms and their glucuronides and sulfates in serum were studied. They found that
after oral dosing of both the parent forms, morin, and its glucuronides, and sulfates
were present in the blood stream and a nonlinear pharmacokinetics was observed
for morin. On the other hand, negligible bioavailability of quercetin is presented as
its glucuronides and sulfates were only detected in the blood and the metabolites
showed linear pharmacokinetics at the two doses studied. The total AUC of parent
form with conjugated metabolites showed that the extent of absorption of morin
was threefold compared to that of quercetin. Hou et al. [18] proved that the fates of
the flavonols were markedly affected by difference in hydroxylation pattern on
B-ring. Xie et al. showed how morin interacted with human serum albumin (HSA).
The interaction has been investigated using fluorescence, Fourier transform,
infrared spectroscopic approaches, and UV absorption [45]. Under the physiolog-
ical condition, there is a specific binding site on HSA for morin, and the binding
affinity was found to be 1.13 ± 0.11 10−5 L Mol−1. The intrinsic fluorescence
of morin was noticeably boosted in the presence of HSA due to excited-state proton
transfer. The level of protonation of the hydroxyl groups played a significant role
during the morin–HSA binding process and it was evident from the fact that with
the increase of the buffer pH from 6.4 to 8.4, binding ability of morin to protein
decreased. The interaction between morin and HSA induced an apparent decline of
the protein a-helix and b-sheet structures [45].
19 Morin and Its Role in Chronic Diseases 457
Morin proves itself effective against many chronic diseases. An increasing number of
studies showed that morin significantly modulate different cell signaling pathways
related to chronic pathophysiological conditions, including gastrointestinal com-
plications, diabetes, cardiovascular disease, cancer, arthritis, neurodegenerative
disease, and several inflammatory diseases. As various oxidative stress-related dis-
orders are led by inflammation, antioxidant and anti-inflammatory activities of morin
play a critical role in the therapeutic process of these disorders. Different studies
prove that morin is effective against several chronic diseases like chronic neurode-
generative disease, chronic cardiovascular pathophysiology, chronic gastrointestinal
pathophysiology, chronic hypersensitivity and immunological disorders, cancer and
several oxidative stress-related chronic pathophysiological disorders, arthritis, dia-
betes, and related pathophysiology. Morin alters the levels of phosphorylated Akt
kinase, Erk1/2, AIF release, cytosolic Bax, and regulated the NF-jB’s nuclear
translocation, and acts against the imbalances in the levels/activities of different
enzymes (e.g., elastase), inflammatory mediators, proinflammatory cytokines,
inflammatory enzymes, glycoproteins, reactive oxygen species, lysosomal acid
hydrolases, and transcription factors (e.g., NF-jB, p65, and AP1) to produce the
desirable effects. Also, different others pathways are involved in morin’s protective
actions and these are described extensively in the following sections.
neuronal death [7]. Zhang et al. investigated the neuroprotective effects of morin on
1-methyl-4-phenylpyridinium ion (MPP+)-induced apoptosis in neuronal differen-
tiated PC12 cells as well as in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine
(MPTP) mouse model of Parkinson disease (PD) where they found that MPP+, in
PC12 cells, induced ROS formation and apoptosis, whereas morin significantly
attenuated the MPP+-induced loss of cell viability, apoptosis, and inhibit ROS
formation. In mice model, morin significantly attenuated the MPTP-induced
nigrostriatal lesions, dopaminergic neuronal death, striatal dopamine depletion, and
permanent behavioral deficits. The results clearly suggest the neuroprotective
actions of this unique molecule both in vitro and in vivo and indicate the possibility
of being a novel therapeutic agent for the treatment of PD and other neurodegen-
erative diseases [49].
Ammonia is considered as a potent neurotoxin. It has been intensely associated
in the pathogenesis of hepatic encephalopathy. Subash and Subramaniam evaluated
the chronotherapeutic effect of morin, on ammonium chloride (AC)-induced
hyperammonemia in a rat model. Morin significantly ameliorate AC-induced
pathophysiological changes in respect to the circulating levels of urea, ammonia,
hydroperoxides (HP), thiobarbituric acid reactive substances (TBARS), liver
markers [aspartate transaminase (AST), alanine transaminase (ALT), and alka-
linephosphatase (ALP)], superoxide dismutase (SOD), glutathione peroxidase
(GPx), catalase (CAT), reduced glutathione (GSH), and vitamins A, C, and E. The
authors speculated that the chronotherapeutic effect of morin in hyperammonemic
rats might be due to temporal variations of lipid peroxidation and of antioxidants,
urea cycle enzymes, etc., temporal variations of metabolic enzymes involved in the
degradation of morin and the temporal variation in its bioavailability [36].
ethanolic extract of these leaves probably exerts antiarthritic activity after oral
administration and it also has antioxidant properties; combination of the both may
contribute to its beneficial activity [3]. Zeng et al. [48] investigated the effect of
morin on type II collagen-induced arthritis (CIA) in rats and also explored the
underlying molecular mechanisms of synovial angiogenesis. Morin significantly
attenuated arthritic development which is specified by reduction of paw swelling
and arthritis scores. Morin also noticeably reduced the serum levels of proinflam-
matory cytokines, e.g., interleukin (IL)-6 (IL6), tumor necrosis factor-a (TNFa),
etc. Along with that, it increased the level of anti-inflammatory cytokine
interleukin-10 and improved pathological changes of joints as evident from his-
tology. Morin also distinctly inhibited expression of vascular endothelial growth
factor (VEGF), basic fibroblast growth factor, and CD31 in synovial membrane
tissues. It also reduced the serum levels of VEGF in CIA rats. In in vitro study,
morin also significantly inhibited VEGF-induced human umbilical vein endothelial
cells migration and tube formation. These results nicely showed that morin had
antirheumatoid potential which it exerts probably by inhibiting synovial [48].
Sultana and Rasool proved that the combination therapy of morin along with a
NSAID was very effective in suppressing the pathogenesis of rheumatoid arthritis
(RA), against adjuvant-induced arthritis (an experimental model for RA) in rats.
They found that imbalances in the levels/activities of elastase, inflammatory
mediators (TNFa, IL1b, MCP1, VEGF, and PGE2), paw edema, glycoproteins
(hexose and hexosamine), urinary constituents (hydroxyproline and glycosamino-
glycans), reactive oxygen species (LPO and NO), lysosomal acid hydrolases (acid
phosphatase, N-acetylglucosaminidase, b-galactosidase, and cathepsin D),
proinflammatory cytokines (TNFa, IL1b, IL17, IL6, and MCP1), inflammatory
enzymes (iNOS and COX2), RANKL, and transcription factors (NF-jB, p65, and
AP1) were regulated back effectively to near control level by morin and indo-
methacin, which were elevated in case of RA. Their findings were supported by
histopathological and radiological analysis, whereas body weight, bone collagen,
and the antioxidant status [superoxide dismutase (SOD), catalase (CAT), glu-
tathione peroxidase (GPx), glutathione, and ceruloplasmin)] were found to be
decreased in RA and that was restored back by the combinatorial therapy [37]. Gout
is a general systemic joint disorder where hyperuricemias is a hallmark of gout [3].
A serum uric acid level above 9 mg dL−1 is considered as gouty arthritis [6].
Pathological manifestation of gout follows over production or decreased excretion
of uric acid (purine metabolic end product) [26]. Urate–anion transporter (URAT1)
in the brush border membrane of the proximal tubule in kidney is the main
transporter involved in the maintenance of serum uric acid level by reabsorbing the
urate from the lumen to the cytosol in kidney tubules [9, 14]. Different drugs
including xanthine oxidase inhibitors (e.g., allopurinol), inhibitors of urate reab-
sorption at proximal renal tubule-like probenecid, benzbromarone, etc. are being
used in the treatment of gout. However, some undesirable side effects like hepa-
totoxicity are associated with the benzbromarone and other agents having
hypouricemic activity [14]. Certain natural herbs were reported to have the xanthine
oxidase inhibitor activity along with other types of mechanisms and those are
460 K. Sinha et al.
Cardiovascular diseases (CVD) are the major cause of chief mortality worldwide due
to its complicated nature. Among CVD disorder, MI is a major one. If there is
imbalance between the coronary supply and its myocardial demand, then myocardial
infarction takes place and it causes necrosis of the myocardial tissue. Morin shows
cardiovascular protection in isoproterenol (ISO)-induced myocardial infarction in
rats and this protection is attributed to its free radical scavenging activity by the
polyphenolic group [2]. It also shows significant beneficial effect on lipid profiles,
blood pressure, and serum glucose levels from the high-fat diet-induced hyperten-
sive rats. ISO, which is a synthetic catecholamine, has been documented to produce
severe stress in the myocardium, resulting in myocardial infarction, if it is admin-
istered in supramaximal doses. Cardiac necrosis due to the administration of ISO
includes increased oxygen consumption, calcium overload and accumulation,
increased myocardial cAMP levels, alterations of membrane permeability, and
increase in lipid peroxides level [2]. Al-Numair et al. reported that morin pretreat-
ment (20, 40, and 80 mg/kg, respectively) daily for a period of 30 days decreases
significantly the activities of cardiac marker enzymes such as lactate dehydrogenase,
creatine kinase, aspartate transaminase and creatine kinase-MB, membrane bound
enzymes (such as calcium-dependent adenosine triphosphatase), sodium
potassium-dependent adenosine triphosphatase, and magnesium-dependent adeno-
sine triphosphatase, in serum. Calcium-dependent adenosine triphosphatase and
magnesium-dependent adenosine triphosphatase were increased, whereas the
activity of sodium potassium-dependent adenosine triphosphatase was found to
decrease in the heart. At the same time it also showed a significant decrease in
glycoprotein (hexosamine, fucose, hexose, and sialic acid) levels in serum and heart.
ISO administration disrupts the redox balance and produces myocardial infarction
via free radical-mediated b-adreno-receptor mechanism. However, positive alter-
ations of these biochemical parameters were successfully achieved with morin
pretreatment daily for a period of 30 days. From their studies, the authors concluded
that morin has a protective role in ISO-induced MI in rats and the observed effects
might be due to the free radical scavenging, antioxidant, and membrane-stabilizing
19 Morin and Its Role in Chronic Diseases 461
properties of morin [2]. These beneficial effects of morin are due to its multitude of
biological function such as antioxidant, anti-inflammatory, and free radical scav-
enging activity. Another group of investigators also conducted a study to prove the
cardio-protective benefits of this flavonoid in ISO-induced myocardial infarcted rats.
A significant increase in the levels of cardiac markers was detected in ISO-induced
myocardial infarcted rats although morin pretreated animals could regulate the
abnormalities in electrocardiograph and biomarkers. Results also showed an
increased lipid peroxidation product in ISO-induced myocardial infarcted rats. The
animals, pretreated with morin, however, showed reduction of lipid peroxidation.
Histopathological studies supported all these observations as pretreatment with
morin-inhibited myocardial damage. Combining, the results of the above-mentioned
studies proved that the pretreatment of morin exhibits protective effect and are
rational to understand its beneficial effects on cardio-protection against myocardial
injury. The results also indicate the cardio-protective ability of morin [31]. However,
till now, the molecular mechanism of its protective actions is not crystal clear, and it
requires more studies to prove its inherent molecular mechanisms.
The protective effect of morin against deoxycorticosterone acetate (DOCA)-
induced hypertension was recently investigated in male Wistar rats. DOAC salt
hypertensive rats showed considerably increased systolic and diastolic blood pres-
sure in association with considerably increased systolic and diastolic blood, AST,
ALP, ALT, GGT, urea, uric acid, and creatinine levels in the plasma. However,
morin effectively lowered all the enzymes’ level up to that of the control. The study
indicates antihypertensive effect of morin [32]. In vitro studies have established that
oxidized low-density lipoprotein (ox-LDL) has increased atherogenicity compared
to native LDL. Upon oxidative modification of LDL, alteration in its structure allows
LDL to be taken up by scavenger receptors on smooth muscle, macrophage, and
endothelial cells. This lead to the formation of lipid-laden foam cells which is the
hallmark of primary atherosclerotic lesions. Naderi et al. found that morin signifi-
cantly inhibits in vitro LDL oxidation. Thus, they proved that morin would probably
be helpful to prevent atherosclerosis [28]. In a study, Wu et al. demonstrated that
morin hydrate significantly reduced the tissue necrosis in post-ischemic and reper-
fused rabbit hearts by >50 %. They showed that, besides scavenging oxyradicals,
morin hydrate discreetly inhibits a free radical generating enzyme xanthine oxidase,
from the ischemic endothelium. However, they also speculate that morin hydrate
may chelate some metal ions which help in further oxyradical formation and thus
inhibiting the process of oxidative stress generation [44].
b Fig. 19.2 Effect of morin on gastric mucosa in IND-induced gastric injury. Control: vehicle
treatment alone; IND: IND treatment alone; morin + IND: treatment with morin IND; Morin:
morin treatment alone. a Open stomach showing injured mucosa. Note that the injury (in the form
of reddish black ulcers of different sizes) is highest in the IND (red arrows) and morin almost
completely prevented the ulceration. b Sections of gastric mucosa were stained with hematoxylin–
eosin. The IND group showed marked changes with outward mucosal damage (red arrow),
presence of inflammatory exudates (green arrows), extensive vasocongestion (black arrows), and
damaged submucosa (blue arrow) [Reprinted from Biochim. Biophys. Acta [General Subjects]
1850:769–783; with the permission from Elsevier; License Number: 3764180672838]
synthesis of most important cytokine, IL-1b, decreased in the NOS and free radicals
involved in the inflammatory cascade are believed to be the factors involved in the
anti-inflammatory activity of morin.
19 Morin and Its Role in Chronic Diseases 465
Noor et al. [29] showed that morin hydrate inhibits amyloid formation by human
islet amyloid polypeptide (IAPP, amylin) and disaggregates preformed IAPP
amyloid fibers that were evident from transmission electron microscopy (TEM) and
right-angle light scattering. IAPP is responsible for type-2 diabetes-related islet
amyloid formation and is also evident in islet cell transplants leading to graft
failure. Human IAPP have fewer inhibitors and it is extremely amyloidogenic.
However, due to the specific substitution pattern on the B-ring, morin hydrate
signifies a novel type of IAPP amyloid inhibitor [29]. Vanitha et al. [41] found that
morin significantly reduced the blood glucose and enhanced the serum insulin
levels in experimentally induced type I diabetic rats. Morin dose dependently
reduced glucose-6-phosphatase, fructose-1,6-bisphosphatase, hexokinase, and
glucose-6-phosphate dehydrogenase activities in liver. It significantly protected
pancreatic islets’ overall morphology as well as preserve insulin-positive cells in
diabetic rats. From the study it is evident that morin might be advantageous in the
treatment of diabetes and regulation of carbohydrate metabolic enzyme activities. It
is of prime importance in the pathophysiological conditions [41]. Abuohashish et al.
[1] found the putative advantageous effect of morin on diabetic osteopenia in rats
which they suggested might be due to both the anti-inflammatory and antioxidant
properties. They found that morin is particularly effective in bone metabolic dis-
order which is a major chronic problem in age-related disease. In case of diabetic
rats significant bone loss was observed at the level of bone turnover parameters
including osteocalcin (OC), bone alkaline phosphatase (bALP), and collagen type 1
cross-linked C-telopeptide (CTX). However, the study showed that morin treatment
obviously attenuated these elevations in those parameters [1]. A significant
impairment in trabecular bone microarchitecture, density, and other morphometric
parameters have been detected by performing bone micro-CT scan of diabetic rats,
and those were efficiently ameliorated by morin treatment. Besides, in diabetic rats,
serum levels of glucose, TBARS, IL-1b, IL-6, and TNF-a were significantly ele-
vated, while that of insulin and GSH was decreased. These changes were bring back
to normal after 5 weeks morin treatment, suggesting the protective effect of morin
against diabetic-induced osteopenia [1].
leukemia (RBL)-2H3 cells and bone marrow-derived mast cells (BMMCs) along
with the inhibition of p38, Akt and the MAP kinases, ERK1/2, JNK, and Fyn
kinase. With further investigations, their results suggest that the morin suppresses
the IgE-mediated allergic response by principally inhibiting Fyn kinase in mast cells
[23]. Findings of Fang et al. [10] indicated that morin might have the ability to
regulate immune response through modulating the cytokine profiles exhibited in
chronic immunotoxic pathophysiology. They have shown that morin and its
derivatives (sulfates/glucuronides) were effective on LPS-activated RAW 264.7
cells (macrophages) in reducing NO, TNF-a, and IL-12 production. Moreover,
phagocytic activities of the peripheral blood cells were significantly lowered in the
morin-treated cells in respect to control. These lowering in the NO production and
reduced macrophage phagocytic activities corresponded to LPS-resistant state is
very important to treat various chronic autoimmune diseases [10].
Recently, attention has been given on the anti-cancer activity of morin in various
kinds of cancers. For example, it exerted protective effect on chemically produced
rat tongue carcinogenesis [20] and inhibited phorbol ester-induced transformation
of rat hepatocytes.[19]. Besides, by inhibiting the lipoxygenase pathway, this
molecule could inhibit the peroxisome-proliferated activator receptor-induced ker-
atinocyte differentiation [39]. The inhibitory activity of this molecule was reported
during the release of inflammatory cytokines (such as IL-8, IL-6, and TNF) from
mast cells [21]. In an in vivo study, morin hydrate showed anticancer activity in
cancer models (like inhibit the growth of COLO205 cells in nude mice) [8]. The
transcription factor NF-jB is known to be involved in various kinds of cell pro-
liferation, cell survival, tumorigenesis, and inflammation. Induction of NF-jB
activation pathway induced by TNF, ceramide, lipopolysaccharide, IL-1, phorbol
12-myristate 13-acetate, and H2O2 was reported to be suppressed by morin. The
process involved the inhibition of IjB (inhibitory subunit of NF-jB) kinase that
leads to suppression of phosphorylation and degradation of IjBa and consequently
nuclear translocation of p65 occurs. NF-jB-dependent reporter gene expression
[activated by TNF receptor (TNFR), TNF, TNFR-associated factor 2,
TNFR1-associated death domain, NF-jB-inducing kinase, IǩB kinase, and the p65
subunit of NF-jB] was also reported to be inhibited by morin hydrate. Besides, it
could also downregulate the NF-jB-related products that are involved in the cell
survival (i.e., inhibitor of apoptosis proteins 1 & 2, survivin, X-chromosome-linked
IAP, and BcL-xL), invasion (matrix metalloproteinase-9), and proliferation (cyclin
D1 and cyclooxygenase-2) [14].
Zeng et al. in their study demonstrated that morin hydrate acts as a broad-
spectrum antioxidant as it scavenges both xanthine oxidase/hypoxanthine-generated
19 Morin and Its Role in Chronic Diseases 467
19.4 Conclusions
From the diverse studies reviewed in this article, morin emerged as a useful natural
flavone in the management of different chronic pathophysiological conditions.
Despite these direct pharmacological activities, morin could help to detect param-
eters related to severe chronic diseases. It could also be used as an excellent and
novel pathological detection tool. Recently, fluorescence-based, time saving, stain
with morin hydrate has been developed and it stains phospho-proteins in
one-dimensional SDS-PAGE where Al3+ was used as a ‘‘fixed bridge.’. With the
help of this novel quantification method based on kinetic measurement of the
fluorescence decrease of Al3+–morin complex it can be used to determine the
bisphosphonate content in plasma samples. This method would upkeep research on
the development of drug delivery systems for the related bisphosphonates.
However, despite the in vivo and in vitro studies, trying to elucidate the mecha-
nisms of action morin, in future, more studies are required to clearly understand the
precise molecular mechanism with great depth. Detailed preclinical studies and its
clinical experimentations are also extremely needed to provide a basis for potential
expediency of this gift of nature, morin, in the treatment and mitigation of chronic
diseases.
References
1. Abuohashish HM, Al-Rejaie SS, Al-Hosaini KA, Parmar MY, Ahmed MM (2013) Alleviating
effects of morin against experimentally-induced diabetic osteopenia. Diabetol Metab Syndr 5:1
2. Al-Numair KS, Chandramohan G, Alsaif MA (2012) Pretreatment with morin, a flavonoid,
ameliorates adenosine triphosphatases and glycoproteins in isoproterenol-induced myocardial
infarction in rats. J Nat Med 66:95–101
3. Amo-Barimah A, Woode E, Boakye-Gyasi E, Ainooson G, Abotsi W (2010) Antiarthritic and
antioxidant effects of the leaf extract of Ficus exasperata P. Beauv. (Moraceae). Pharmacognosy
Res 2:89–97.
4. Basile A, Sorbo S, Giordano S, Ricciardi L, Ferrara S, Montesano D, Cobianchi RC,
Vuotto M, Ferrara L (2000) Antibacterial and allelopathic activity of extract from Castanea
sativa leaves. Fitoterapia 71:S110–S116
5. Bradley J-C, Lang AS, Williams AJ, Curtin E (2011) ONS open melting point collection.
Available from Nature Precedings. http://dx.doi.org/10.1038/npre.2011.6229.1
6. Campion EW, Glynn RJ, Delabry LO (1987) Asymptomatic hyperuricemia. Risks and
consequences in the Normative Aging Study. Am J Med 82:421–426
7. Campos-Esparza MR, Sanchez-Gomez MV, Matute C (2009) Molecular mechanisms of
neuroprotection by two natural antioxidant polyphenols. Cell Calcium 45:358–368
8. Chen Y-C, Shen S-C, Chow J-M, Ko CH, Tseng S-W (2004) Flavone inhibition of tumor
growth via apoptosis in vitro and in vivo. Int J Oncol 25:661–670
9. Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, Jutabha P, Cha SH, Hosoyamada M,
Takeda M, Sekine T, Igarashi T (2002) Molecular identification of a renal urate–anion
exchanger that regulates blood urate levels. Nature 417:447–452
19 Morin and Its Role in Chronic Diseases 469
10. Fang S-H, Hou Y-C, Chang W-C, Hsiu S-L, Chao P-DL, Chiang B-L (2003) Morin
sulfates/glucuronides exert anti-inflammatory activity on activated macrophages and decreased
the incidence of septic shock. Life Sci 74:743–756
11. Galvez J, Coelho G, Crespo M, Cruz T, Rodríguez-Cabezas M, Concha A, Gonzalez M,
Zarzuelo A (2001) Intestinal anti-inflammatory activity of morin on chronic experimental
colitis in the rat. Aliment Pharmacol Ther 15:2027–2039
12. Gladding PA, Webster MW, Farrell HB, Zeng IS, Park R, Ruijne N (2008) The antiplatelet
effect of six non-steroidal anti-inflammatory drugs and their pharmacodynamic interaction
with aspirin in healthy volunteers. Am J Cardiol 101:1060–1063
13. Goldstein RS, Schnellmann RG (1996) Toxic responses of the kidney. In: Klaassen CD, (ed.),
Casarett and Doull’s toxicology: the basic science of poisons, 5th edn. Mc Graw Hill, Kansas,
pp 417–442
14. Gopal JV (2013) Morin hydrate: botanical origin, pharmacological activity and its
applications: a mini-review. Pharmacogn J 5:123–126
15. Gottlieb M, Leal-Campanario R, Campos-Esparza MR, Sánchez-Gómez MV, Alberdi E,
Arranz A, Delgado-García JM, Gruart A, Matute C (2006) Neuroprotection by two
polyphenols following excitotoxicity and experimental ischemia. Neurobiol Dis 23:374–386
16. Grisham MB (1992) A comparative analysis of two models of colitis in rats. Gastroenterology
102:1524–1534
17. Hogaboam C, Jacobson K, Collins S, Blennerhassett M (1995) The selective beneficial effects
of nitric oxide inhibition in experimental colitis. Am J Physiol Gastrointest Liver Physiol 268:
G673–G684
18. Hou Y, Chao P, Ho H, Wen C, Hsiu S (2003) Profound difference in pharmacokinetics
between morin and its isomer quercetin in rats. J Pharm Pharmacol 55:199–203
19. Hsiang C-Y, Wu S-L, Ho T-Y (2005) Morin inhibits 12-O-tetradecanoylphorbol-
13-acetate-induced hepatocellular transformation via activator protein 1 signaling pathway
and cell cycle progression. Biochem Pharmacol 69:1603–1611
20. Kawabata K, Tanaka T, Honjo S, Kakumoto M, Hara A, Makita H, Tatematsu N, Ushida J,
Tsuda H, Mori H (1999) Chemopreventive effect of dietary flavonoid morin on chemically
induced rat tongue carcinogenesis. Int J Cancer 83:381–386
21. Kempuraj D, Madhappan B, Christodoulou S, Boucher W, Cao J, Papadopoulou N,
Cetrulo CL, Theoharides TC (2005) Flavonols inhibit proinflammatory mediator release,
intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast
cells. Br J Pharmacol 145:934–944
22. Kim JM, Lee EK, Park G, Kim MK, Yokozawa T, Yu BP, Chung HY (2010) Morin
modulates the oxidative stress-induced NF-jB pathway through its anti-oxidant activity. Free
Radical Res 44:454–461
23. Kim JW, Lee JH, Hwang BY, Mun SH, Ko NY, Kim DK, Kim B, Kim HS, Kim YM,
Choi WS (2009) Morin inhibits Fyn kinase in mast cells and IgE-mediated type I
hypersensitivity response in vivo. Biochem Pharmacol 77:1506–1512
24. Kuhnau J (1976) Flavonoids. A class of semi-essential food components: Their role in human
nutrition. In: World review of nutrition and dietetics. World Rev Nutr Diet 24:117–191
25. Lian T-W, Wang L, Lo Y-H, Huang I-J, Wu M-J (2008) Fisetin, morin and myricetin attenuate
CD36 expression and oxLDL uptake in U937-derived macrophages. Biochim Biophys Acta
Mol Cell Biol Lipids 1781:601–609
26. Lioté F (2003) Hyperuricemia and gout. Curr Rheumatol Rep 5:227–234
27. Middleton E, Kandaswami C, Theoharides TC (2000) The effects of plant flavonoids on
mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev
52:673–751
470 K. Sinha et al.
28. Naderi GA, Asgary S, Sarraf-Zadegan N, Shirvany H (2003) Anti-oxidant effect of flavonoids
on the susceptibility of LDL oxidation. Mol Cell Biochem 246:193–196
29. Noor H, Cao P, Raleigh DP (2012) Morin hydrate inhibits amyloid formation by islet amyloid
polypeptide and disaggregates amyloid fibers. Protein Sci 21:373–382
30. Park C, Lee WS, Go S-I, Nagappan A, Han MH, Hong SH, Kim GS, Kim GY, Kwon TK,
Ryu CH (2014) Morin, a flavonoid from Moraceae, induces apoptosis by induction of BAD
protein in human leukemic cells. Int J Mol Sci 16:645–659
31. Pogula BK, Maharajan MK, Oddepalli DR, Boini L, Arella M, Sabarimuthu DQ (2012) Morin
protects heart from beta-adrenergic-stimulated myocardial infarction: an electrocardiographic,
biochemical, and histological study in rats. J Physiol Biochem 68:433–446
32. Prahalathan P, Kumar S, Raja B (2012) Effect of morin, a flavonoid against DOCA-salt
hypertensive rats: a dose dependent study. Asian Pac J Trop Biomed 2:443–448
33. Salzman A, Denenberg AG, Ueta I, O’Connor M, Linn SC, Szabó C (1996) Induction and
activity of nitric oxide synthase in cultured human intestinal epithelial monolayers. Am J
Physiol Gastrointest Liver Physiol 270:G565–G573
34. Singh MP, Jakhar R, Kang SC (2015) Morin hydrate attenuates the acrylamide-induced
imbalance in antioxidant enzymes in a murine model. Int J Mol Med 36:992–1000
35. Sinha K, Sadhukhan P, Saha S, Pal PB, Sil PC (2015) Morin protects gastric mucosa from
nonsteroidal anti-inflammatory drug, indomethacin induced inflammatory damage and
apoptosis by modulating NF-jB pathway. Biochim Biophys Acta Gen Subj 1850:769–783
36. Subash S, Subramanian P (2012) Chronotherapeutic effect of morin in experimental chronic
hyperammonemic rats. Int J Nutr Pharmacol Neurol Dis 2:266
37. Sultana F, Rasool M (2015) A novel therapeutic approach targeting rheumatoid arthritis by
combined administration of morin, a dietary flavanol and non-steroidal anti-inflammatory drug
indomethacin with reference to pro-inflammatory cytokines, inflammatory enzymes, RANKL
and transcription factors. Chem Biol Interact 230:58–70
38. Surh Y-J (2004) Transcription factors in the cellular signaling network as prime targets of
chemopreventive phytochemicals. Cancer Res Treat 36:275
39. Thuillier P, Brash A, Kehrer J, Stimmel J, Leesnitzer L, Yang P, Newman R, Fischer S (2002)
Inhibition of peroxisome proliferator-activated receptor (PPAR)-mediated keratinocyte
differentiation by lipoxygenase inhibitors. Biochem J 366:901–910
40. Travis S, Jewell D (1994) Salicylates for ulcerative colitis—their mode of action. Pharmacol
Ther 63:135–161
41. Vanitha P, Uma C, Suganya N, Bhakkiyalakshmi E, Suriyanarayanan S, Gunasekaran P,
Sivasubramanian S, Ramkumar K (2014) Modulatory effects of morin on hyperglycemia by
attenuating the hepatic key enzymes of carbohydrate metabolism and b-cell function in
streptozotocin-induced diabetic rats. Environ Toxicol Pharmacol 37:326–335
42. Venkatesan R, Sadiq AM, Kumar JS, Lakshmi GR, Vidhya R (2010) Effect of Morin on
mercury chloride induced nephrotoxicity. Ecoscan 4:193–196
43. Wijeratne SS, Abou-Zaid MM, Shahidi F (2006) Antioxidant polyphenols in almond and its
coproducts. J Agric Food Chem 54:312–318
44. Wu T-W, Fung K-P, Zeng L-H, Wu J, Hempel A, Grey AA, Camerman N (1995) Molecular
properties and myocardial salvage effects of morin hydrate. Biochem Pharmacol 49:537–543
45. Xie M-X, Long M, Liu Y, Qin C, Wang Y-D (2006) Characterization of the interaction
between human serum albumin and morin. Biochim Biophys Acta Gen Subj 1760:1184–1191
46. Yu Z, Fong WP, Cheng CH (2007) Morin (3,5,7,2′,4′-pentahydroxyflavone) exhibits potent
inhibitory actions on urate transport by the human urate anion transporter (hURAT1)
expressed in human embryonic kidney cells. Drug Metab Dispos 35:981–986
47. Zeng L, Rootman D, Burnstein A, Wu J, Wu T (1998) Morin hydrate: a better protector than
purpurogallin of corneal endothelial cell damage induced by xanthine oxidase and SIN-1. Curr
Eye Res 17:149–152
19 Morin and Its Role in Chronic Diseases 471
48. Zeng N, Tong B, Zhang X, Dou Y, Wu X, Xia Y, Dai Y, Wei Z (2015) Antiarthritis effect of
morin is associated with inhibition of synovial angiogensis. Drug Dev Res 76:463–73
49. Zhang Z-T, Cao X-B, Xiong N, Wang H-C, Huang J-S, Sun S-G, Wang T (2010) Morin exerts
neuroprotective actions in Parkinson disease models in vitro and in vivo. Acta Pharmacol Sin
31:900–906
Chapter 20
Ellagic Acid and Its Role in Chronic
Diseases
Abstract Ellagic acid is a natural anti-oxidant phenol found in numerous fruits and
vegetables, in particular pomegranate, persimmon, raspberry, black raspberry,
strawberry, peach, plumes, nuts (walnuts, almonds), and wine. The anti-proliferative
and anti-oxidant properties of ellagic acid have prompted research into its potential
health benefits. The aim of this chapter will be to summarize potential benefits of
ellagic acid supplementation in chronic diseases.
20.1 Introduction
A diet rich in polyphenols, which are found in fruits and vegetables, is strongly
associated with a reduced risk for developing chronic diseases such as cancer and
cardiovascular disease [1]. Ellagic acid is a natural phenol anti-oxidant found in
numerous fruits and vegetables. Ellagic acid is a dimeric derivative of gallic acid
and rarely occurs free in diet crops, but usually occurs in food products conjugated
with glycoside moiety (glucose, xylose) or forms part of ellagitannins (polymeric
molecules) [2, 3]. These compounds usually occur in fruits (pomegranates, per-
simmon, raspberries, black raspberries, strawberries, peach, plumes), nuts (walnuts,
almonds), vegetables, and wine. Ellagitannins are the bioactive polyphenols present
in pomegranate; however, they are not absorbed intact by the human gut, but they
can be hydrolyzed to ellagic acid by colonic gastrointestinal flora [3, 4]. The
anti-proliferative and anti-oxidant properties of ellagic acid have prompted research
into its potential health benefits. The aim of this chapter will be to summarize
potential benefits of ellagic acid supplementation.
The anti-cancer effect of ellagic acid has been studied in many human cancer cell
lines including those of skin, esophageal, and colon cancer where it exhibited
anti-proliferative activity, with the ability to cause cell cycle arrest and induce
476 G. Derosa et al.
apoptosis [8]. Ellagic acid takes part in various DNA maintenance reactions pre-
venting genomic instability which otherwise leads to cancer [9]. Ellagic acid has an
anti-proliferative effect and induces apoptosis via a mitochondrial pathway in
Caco-2 cells, an in vitro model for colon cancer, without interfering with the normal
colon cells [3]. In particular, a study exploring the activity of ellagic acid
demonstrated that it could induce apoptosis in 1,2-dimethyl hydrazine (DMH)-
induced colon carcinoma and participate in a wide range of DNA maintenance
reactions that prevent genomic instability [8]. Ellagic acid prevents PI3K/Akt
activation that, in turn, results in the modulation of its downstream Bcl-2 family
proteins [8] involved in the activation of the intrinsic apoptotic pathway [3]. Bax
expression and caspase-3 activation were noted after ellagic acid supplementation
leading to elevation of cytochrome c levels and finally cell death [3].
It has been reported that the gradual build-up of advanced glycation end products
(AGEs) in body tissues is a major contributor to many progressive diseases
including diabetic complications [16] and Alzheimer’s disease [17, 18].
AGE-modified plasma proteins could bind to AGE receptors (RAGE) on the sur-
face of the cell, activate cell signaling, and lead to the production of reactive oxygen
species and inflammatory factors. Moreover, proteins on the extracellular matrix
crosslink with other matrix components, leading to a loss in their function.
As mentioned above, pomegranate fruit, which is a rich source of phenolics, in
particular ellagitannins, showed anti-oxidant and anti-inflammatory effects. Taking
into account the effects on AGE, pomegranate fruit juice and extracts have been
shown to exert neuro-protective effects by ameliorating symptoms of Alzheimer’s
disease potentially caused by the abnormal accumulation of AGEs in the brain [19].
This finding was further supported by Rojanathammanee et al. [20], who
determined whether a dietary intervention with ellagic acid could attenuate
microgliosis in a rat model of Alzheimer’s disease. Three months of pomegranate
feeding lowered TNF-α concentrations in brain, and lowered nuclear factor of
activated T-cell (NFAT) transcriptional activity compared with controls.
Immunocytochemistry showed that pomegranate, but not control-fed mice, had
attenuated microgliosis and amyloid β plaque deposition, involved in Alzheimer’s
disease.
Ellagic acid seems to play an anti-diabetic activity through the action on β-cells of
pancreas, stimulating insulin secretion and decreasing glucose intolerance. This
effect of ellagic acid was reported by Fatima et al. [21]. Treatment with Emblica
officinalis extract, rich in ellagic acid, has been reported to result in a significant
decrease in the fasting blood glucose in a dose- and time-dependent manner in
diabetic rats. Ellagic acid significantly increased serum insulin in diabetic rats in a
dose-dependent manner. Insulin-to-glucose ratio was also increased by Emblica
officinalis treatment. Immunostaining of pancreas showed that Emblica officinalis
extract (250 mg/kg) increased β-cell size, but a higher dose of 500 mg/kg increased
β-cells number in diabetic rats. Moreover, Emblica officinalis extract significantly
increased plasma total anti-oxidants and liver GSH and TBARS. Elevation in
glucose tolerance by ellagic acid suggested that ellagic acid probably works by
stimulating insulin secretion from pancreatic β-cells. These results infer that ellagic
acid may exert anti-diabetic activity through the action on β-cells of pancreas
resulting in an increase in β-cell size and number, increasing anti-oxidant status,
decreasing blood glucose, increasing serum insulin, and β-cell morphology, and
morphometry [21].
478 G. Derosa et al.
20.4 Conclusions
Ellagic acid seems to be a very promising agent for the treatment of chronic
diseases, especially ulcerative colitis, Cronh’s disease, Alzheimer’s disease, and
diabetes. Ellagic acid also seems to have hepatoprotective and anti-cancer proper-
ties. The anti-diabetic potential of ellagic acid makes it a potentially interesting
alternative to traditional glucose-lowering medications and a strong candidate for
diabetic drug research. However, detailed toxicity evaluations need to be performed
before any therapeutic application in humans could be suggested. Proof-of-concept
safety and efficacy trials are also warranted to verify the interesting preclinical
findings. Further studies are needed to see if the very good results observed in
animal will be confirmed in humans.
References
10. Devipriya N, Sudheer AR, Menon VP (2007) Dose-response effect of ellagic acid
oncirculatory antioxidants and lipids during alcohol-induced toxicity in experimental rats.
Fundam Clin Pharmacol 21:621–630
11. Singh K, Khanna AK, Chander R (1999) Hepatoprotective activity of ellagic acid carbon
tetrachloride induced hepatotoxicity in rats. Indian J Exp Biol 37:1025–1026
12. Yu YM, Chang WC, Wu CS, Chiang SY (2005) Reduction of oxidative stress and apoptosis in
hyperlipidemic rabbits by ellagic acid. J Nutr Biochem 16:675–681
13. Pathak RK, Baunthiyal M, Taj G, Kumar A (2014) Virtual screening of natural inhibitors to
the predicted HBx protein structure of Hepatitis B Virus using molecular docking for
identification of potential lead molecules for liver cancer. Bioinformation 10:428–435
14. Reddy BU, Mullick R, Kumar A, Sudha G, Srinivasan N, Das S (2014) Small molecule
inhibitors of HCV replication from Pomegranate. Sci Rep 4:1–10
15. Ajala OS, Jukov A, Ma CM (2014) Hepatitis C virus inhibitory hydrolysable tanninsfrom the
fruits of Terminalia chebula. Fitoterapia 99:117–123
16. Ulrich P, Cerami A (2001) Protein glycation, diabetes, and aging. Recent Prog Horm Res
56:1–21
17. Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery T, Zhao L, Nagashima M, Morser J,
Migheli A, Nawroth P, Stern D, Schmidt AM (1996) RAGE and amyloid-beta peptide
neurotoxicity in Alzheimer’s disease. Nature 382(6593):685–691
18. Srikanth V, Maczurek A, Phan T, Steele M, Westcott B, Juskiw D, Münch G (2011) Advanced
glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol Aging 32
(5):763–777
19. Liu W, Ma H, Frost L, Yuan T, Dain JA, Seeram NP (2014) Pomegranate phenolics inhibit
formation of advanced glycation endproducts by scavenging reactive carbonyl species. Food
Funct 5(11):2996–3004
20. Rojanathammanee L, Puig KL, Combs CK (2013) Pomegranate polyphenols and extract
inhibit nuclear factor of activated T-cell activity and microglial activation in vitro and in a
transgenic mouse model of Alzheimer disease. J Nutr 143(5):597–605
21. Fatima N, Hafizur RM, Hameed A, Ahmed S, Nisar M, Kabir N (2015) Ellagic acid in
Emblica officinalis exerts anti-diabetic activity through the action on β-cells of pancreas. Eur J
Nutr. doi:10.1007/s00394-015-1103-y
Author Index
A M
Afaq, Farrukh, 213 Maffioli, Pamela, 173, 473
Amin, Shantu G., 375 Mancha-Ramirez, Anna M., 75
Ammon, H.P.T., 291 Monisha, B. Anu, 47
Motiwala, Hashim F., 329
B Moudgil, Kamal D., 267
Baer-Dubowska, Wanda, 131
Baggioni, Alessandra, 27 N
Basu, Pritha, 155 Na, Hye-Kyung, 185
Behera, Amit K., 435 Natesh, Nagashayana, 435
C P
Chin, Kok-Yong, 97 Pal, Harish C., 213
Cicero, Arrigo F.G., 27 Pandey, Manoj K., 375
Cohen, Mark S., 329 Pang, Kok-Lun, 97
Pearlman, Ross L., 213
D Prasad, Ram, 245
Derosa, Giuseppe, 173, 473 Priyadarsini, K.I., 1
G Q
Ghosh, Jyotirmoy, 453 Qiao, Liang, 397
K S
Karelia, Deepkamal, 375 Sahebkar, Amirhossein, 173, 473
Katiyar, Santosh K., 245 Sethi, Gautam, 419
Kumar, Gopinatha Shanmugam, Muthu K., 419
Suresh, 155 Sil, Parames C., 453
Kumar, Niraj, 47 Sinha, Krishnendu, 453
Kundu, Tapas K., 435 Slaga, Thomas J., 75
Kunwar, A., 1 Soelaiman, Ima-Nirwana, 97
Song, Ziwei, 419
L Subramanian, Chitra, 329
Licznerska, Barbara, 131 Surh, Young-Joon, 185
Lu, Hong, 397 Swamy, Mahadeva M., 435
V Y
Venkatesha, Shivaprasad H., 267 Yu, Hanry, 419
W Z
Wang, Jun, 397 Zhou, Yongning, 397