Hepatoprotective Effect of Bisbenzylisoquinoline Alkaloid Tiliamosine From
Hepatoprotective Effect of Bisbenzylisoquinoline Alkaloid Tiliamosine From
net/publication/327904511
CITATIONS READS
21 310
14 authors, including:
All content following this page was uploaded by Sylvester Darvin Sandhaanam on 20 January 2019.
a
Division of Ethnopharmacology, Entomology Research Institute, Loyola College, Chennai, Tamil Nadu, 600 034, India
b
Division of Biotechnology, Entomology Research Institute, Loyola College, Chennai, Tamil Nadu, 600 034, India
c
International Scientific Partnership Programme, King Saud University, Riyadh 11451, Saudi Arabia
d
Addiriyah Chair for Environmental Studies, Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
e
Department of Veterinary Pharmacology and Toxicology, Madras Veterinary College, Chennai, 600 007, India
f
Department of Veterinary Pathology, Madras Veterinary College, Chennai, 600 007, India
Keywords: Non-alcoholic steatohepatitis (NASH) is one of the aggressive forms of non-alcoholic fatty liver disease (NAFLD)
Bisbenzylisoquinoline alkaloids and is a potential risk factor of HCC. This study reports the curative effect of tiliamosine on NASH. Tiliamosine
HepG2 cells was isolated from Tiliacora racemosa Colebr. (Menispermaceae) and its structure was confirmed by studying the
NASH physical and spectroscopic data. The effects of tiliamsoine on lipid accumulation and lipotoxicity were evaluated
NAFLD
using palmitate-oleate induced steatosis in HepG2 cells. The in vivo efficacy of tiliamosine was evaluated using
High fat diet
HFD fed, DEN induced non-alcoholic steatohepatitis Wistar rats. In HepG2 cells, tiliamosine did not affect the
Diethylnitrosamine
cell viability up to 100 μM concentration and showed GI25 value of 264.28 μM. The treatment with tiliamsoine
significantly lowered the ORO concentration by 44.17% and triglyceride accumulation by 69.32% at 50 μM
concentration (P < 0.005). It also reduced the leakage of LDH and transaminases in PO-BSA induced HepG2
cells. The treatment with tiliamsoine significantly decreased the plasma levels of transaminases, phosphatase
and LDH (P < 0.05) in HFD-DEN induced steatohepatitis. The histology and the immunohistochemistry of the
hepatic sections were in accordance with the biochemical findings. Preliminary molecular analysis indicated that
the hepatic FXR expression was upregulated and TNFα expression was downregulated by the treatment with
tiliamsoine. This study provided preliminary evidence on the use of tiliamosine for the treatment of NASH.
Abbreviations: BSA, bovine serum albumin; CMC, carboxymethylcellulose; BBI, bisbenzyl isoquinoline alkaloids; DMSO-d6, deuterated dimethyl sulfoxide; GI,
growth inhibition concentration; HFD, high fat diet; MDA, malondialdehyde; lit.mp., melting point given in the literature; BMI, body mass index; mp, melting point;
MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; NaCl, sodium chloride; NAFLD, non-alcoholic fatty liver disease; NASH, nonalcoholic steato-
hepatitis; ORO, oil red - O; PBS, phosphate buffered saline; POBSA, palmitate, oleate, BSA complex; DEN, diethylnitrosamine; Tc, total cholesterol; Tg, triglyceride
⁎
Corresponding authors at: Division of Ethnopharmacology, Entomology Research Institute, Loyola College, Chennai, Tamil Nadu, 600 034, India.
E-mail addresses: [email protected] (M.G. Paulraj), [email protected] (P. Pandikumar), [email protected] (S. Ignacimuthu).
https://doi.org/10.1016/j.biopha.2018.09.116
Received 10 July 2018; Received in revised form 18 September 2018; Accepted 19 September 2018
0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
964
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Fig. 2. (a) Docked and (b) hydrophobic orientations of tiliamosine and depiction of corresponding amino acid residues with FXRα active site.
Fig. 3. (a) Docked and (b) hydrophobic orientations of WAY-362450 and depiction of corresponding amino acid residues with FXRα active site.
Table 1
Molecular docking of tiliamosine and WAY-362450 with FXRα protein (1OSH).
Ligand Binding amino acid residues Binding Energy Inhibition Constant VDW_HB desolv_energy Ligand efficiency
(kcal/mol) (μM) (kcal/mol)
was calculated using the following formula. 2.3.4. Hepatoprotective effect of tiliamosine on PO-BSA induced
lipotoxicity in HepG2 cells
Cell viability percentage =
Abs570 nm (test material)
× 100 The experiment was carried out according to the previously pub-
Abs570 nm (control) lished procedure [33]. The concentrations of tiliamosine for this study
were fixed between 12.5 and 50 μM, on the basis of MTT assay. Fe-
GI25 value of tilimosine was calculated using linear regression nofibrate was used as the positive control and its dose was fixed as 50
analysis with GraphPad Prism (Version 5.0) and cell viability percen- μM [33]. After 24 h of treatment, the amount of lipids accumulated into
tage. the doses for further studies were kept below its GI25 value. the cells was measured using ORO staining [34]. In another set of
965
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Table 2
Effect of tiliamosine on lipid accumulation and lipotoxicity in PO-BSA induced steatotic HepG2 cells.
Groups % ORO Tg % LDH release MDA level in the cell lysates ALT AST
(mg Tg/ g protein) (nM/mg protein) (IU/L) (IU/L)
Normal control 101.3 ± 2.93a,c 22.31 ± 3.14a 4.84 ± 1.37a 5.77 ± 1.48a 32.76 ± 3.44a 19.22 ± 2.37a
Disease control 167.9 ± 18.65b 79.32 ± 6.52b 46.94 ± 5.23b 29.90 ± 1.58d 101.9 ± 8.48b 48.15 ± 2.49b
Fenofibrate (50 μM) 115.8 ± 10.54a,c 32.76 ± 1.97a,c 3.91 ± 1.38a 12.44 ± 0.60c 38.33 ± 7.22a,c 22.08 ± 1.44a
Tiliamosine(12.5 μM) 121.2 ± 1.14c 34.83 ± 4.82c 18.77 ± 1.33c 11.29 ± 0.48b,c 52.93 ± 2.58c 30.64 ± 1.24c
Tiliamosine (25 μM) 107.9 ± 5.64a,c 27.91 ± 5.60a,c 12.58 ± 1.24c 9.938 ± 0.35b,c 43.31 ± 2.37a,c 28.18 ± 1.44c
Tiliamosine (50 μM) 93.72 ± 4.85a 24.34 ± 3.70a,c 2.98 ± 1.12a 8.94 ± 0.13b 29.74 ± 8.58a 19.66 ± 1.17a
Values indicate mean ± SD of three independent experiments; values having same alphabets in a column did not vary significantly (Tukey’s HSD; P ≤ 0.05).
Table 3
Effect of tiliamosine on the masses of selected organs in animals with HFD-DEN induced non-alcoholic steatohepatitis after four weeks of treatment.
Group Liver Kidney Spleen Adipose
a a a
Normal control 3.14 ± 0.61 0.71 ± 0.20 0.25 ± 0.09 0.79 ± 0.24a,d
Disease control 6.06 ± 1.34b 1.03 ± 0.24a 0.36 ± 0.14a 1.89 ± 0.54b
Sorafenib (60 mg/kg) 3.14 ± 0.49a 0.83 ± 0.05a 0.30 ± 0.04a 1.15 ± 0.19c,d
Tiliamosine (50 mg/kg) 2.58 ± 0.56a 0.67 ± 0.19a 0.26 ± 0.07a 0.54 ± 0.08a
Tiliamosine (100 mg/kg) 2.48 ± 0.80a 0.75 ± 0.31a 0.30 ± 0.06a 0.69 ± 0.19a,c
Values indicate mean ± SD of six animals; Values having same alphabet in a column did not vary significantly (Tukey’s HSD; P ≤ 0.05).
966
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Fig. 5. Representative microphotographs of PO-BSA induced steatotic HepG2 cells treated with tiliamosine.
The cells were treated with the test materials for 24 h and the images were taken after ORO staining at 400× magnification.
MyLab™ Gamma used for the imaging of abdominal organs of the rats
used in the study. The frequency was set at 15 MHz, depth was set at
15 mm. Time Gain Control (TGC) and overall gain were adjusted time
to time to get proper image.
2.3.5.6. End of the study. Feed and water intake were recorded at 9 a.m.
every day and the body weights were measured once a week. The animals
were euthanized under CO2 anesthesia at 28th day after treatment, and blood
was collected through cardiac puncture and stored in EDTA coated tubes.
Plasma was separated from blood after centrifugation at 3500 rpm for 15 min
and stored at 4 °C for biochemical assays. Masses of liver, kidney, spleen and
Fig. 6. Effect of tiliamosine on the body weight of animals with HFD-DEN in- reteroperitoneal adipose tissue were recorded. Liver tissues were trimmed
duced non-alcoholic steatohepatitis after four weeks of treatment. and stored in RNase Later for RNA extraction and fixed in 10% neutral-
Values represent mean ± SD of six animals; Values carrying same alphabet for buffered formalin for histological examination, respectively. Tc (Agappe,
a week did not vary significantly (Tukey’s HSD; P ≤ 0.05). #11403002), Tg (Agappe, #114140002), HDL-c (Agappe, #11414004), ACP
(Accurex, #L02017), ALP (Accurex, #15417), ALT (Accurex, #L04517), AST
2.3.5.5.1. Animal preparation. Rats were anesthetised by an (Accurex, #L06217), GGT (Accurex, #03317), LDH (Accurex, #L11018),
intraperitoneal injection of xylazine (13 mg/kg) and ketamine (87 mg/kg) Albumin (Accurex, #L74716), Total protein (Agappe, #11013003) and Urea
[40]. Immobilisation and anaesthetic effects were noticed after 15 min and (Agappe, #11216001) levels were measured on the same day of euthanasia
the anaesthesia lasted up to 3–4 h post administration of anaesthetics. using commercial spectrophotometric assay kits.
Ventral abdominal areas were shaved, the animals were placed on supine
position on the ultrasound table and the ultrasound gel was applied on the 2.3.5.7. Histopathology and immunohistochemical staining. Neutral buffered,
shaved area before ultrasonography. formalin fixed and paraffin inserted liver tissues were processed and stained
2.3.5.5.2. B-mode ultrasound. The ultrasound scanner, esaote with hematoxylin and eosin. The slides were evaluated for ballooning
Table 4
Effect of tiliamosine on plasma and hepatic lipid profiles in animals with HFD-DEN induced non-alcoholic steatohepatitis after four weeks of treatment.
Group Total cholesterol Triglycerides HDL cholesterol TC/HDL-c Liver lipid
(mg/dL) (mg/dL) (mg/dL) Levels (mg/g liver)
Normal control 49.59 ± 5.22a 75.18 ± 22.32a 68.98 ± 6.61a 0.72 ± 0.13a 31.44 ± 0.83a
Disease control 128.50 ± 27.70b 189.40 ± 58.05b 68.52 ± 6.97a 1.87 ± 0.31b 55.18 ± 11.61b
Sorafenib (60 mg/kg) 98.40 ± 4.65b,d 182.30 ± 8.62b 80.19 ± 11.80a 1.24 ± 0.18a,b 40.95 ± 4.36a,b
Tiliamosine (50 mg/kg) 69.53 ± 28.47a,d 108.20 ± 29.36a 54.07 ± 16.57a 1.33 ± 0.44a,b 33.42 ± 1.72a
Tiliamosine (100 mg/kg) 58.95 ± 7.52a 80.68 ± 35.69a 50.19 ± 21.05a 1.30 ± 0.39a,b 34.60 ± 3.02a
Values indicate mean ± SD of six animals; Values having same alphabet in a column did not vary significantly (Tukey’s HSD; P ≤ 0.05).
967
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Table 5
Effect of tiliamosine on plasma hepatic marker enzymes in animals with HFD-DEN induced non-alcoholic steatohepatitis after four weeks of treatment.
Group ACP ALP ALT AST GGT LDH
a a a a a
Normal control 3.85 ± 0.97 17.37 ± 7.45 32.91 ± 9.43 53.83 ± 7.31 19.19 ± 5.86 334.1 ± 81.26a
Disease control 46.34 ± 13.80b 94.95 ± 10.85b 223.5 ± 66.88b 214.1 ± 67.22b 67.50 ± 9.85b 1711.0 ± 438.6b
Sorafenib (60 mg/kg) 8.15 ± 1.64 51.35 ± 10.58c 81.92 ± 18.78a,c 136.8 ± 23.70c 38.78 ± 6.09c 894.1 ± 213.8a
Tiliamosine (50 mg/kg) 10.90 ± 1.97a 57.67 ± 11.61c 90.60 ± 22.67c 141.6 ± 30.45c 44.55 ± 16.36c 741.4 ± 624.2a
Tiliamosine (100 mg/kg) 9.60 ± 2.62a 50.37 ± 13.69c 96.44 ± 11.27c 131.6 ± 46.09c 47.87 ± 11.73c 654.7 ± 257.6a
Values (in IU/L) indicate mean ± SD of six animals; Values having same alphabet in a column did not vary significantly (Tukey’s HSD; P ≤ 0.05).
Table 6
Effect of tiliamosine on plasma protein and urea levels in animals with HFD-DEN induced non-alcoholic steatohepatitis after four weeks of treatment.
Group Albumin Total protein Albumin/ Urea
(g/dL) (g/dL) Total Protein (mg/dL)
Normal control 1.88 ± 0.09a 6.53 ± 0.53a 0.29 ± 0.03a 15.18 ± 2.86a
Disease control 1.52 ± 0.13b 7.55 ± 1.35a 0.20 ± 0.02b 52.85 ± 2.09b
Sorafenib (60 mg/kg) 1.99 ± 0.14a 7.20 ± 0.88a 0.27 ± 0.01a 25.66 ± 4.50c
Tiliamosine (50 mg/kg) 1.87 ± 0.10a 6.75 ± 0.07a 0.27 ± 0.01a 20.18 ± 6.08a,c
Tiliamosine (100 mg/kg) 1.78 ± 0.31a 6.81 ± 0.46a 0.26 ± 0.06a 27.28 ± 8.15c
Values indicate mean ± SD of six animals; Values having same alphabet in a column did not vary significantly (Tukey’s HSD; P ≤ 0.05).
degeneration, apoptosis, fatty changes and MNC infiltration. Liver cell 3. Results
proliferation was semi-quantitatively assessed by immunostaining using
proliferating cell nuclear antigen (PCNA). Briefly tissue sections after 3.1. Phytochemistry
regular dehydration and rehydration, were incubated with 3% H2O2 for
15 min. Then, they were heated using a steamer for 20 m in 10 mM sodium Tiliamosine (Fig. 1) was isolated form aerial parts of T. racemosa as
citrate (pH 6.0) buffer to retrieve antigen. The routine biotin-streptavidin pale brown amorphous powder. It analysed for C36H36N2O6 and the
immunohistochemical method consisted of sequential incubations in goat or ESI-MS gave [M + H]+ at m/z 593. It gave positive Drangendorff’s test
horse serum blocking solution, and streptavidin conjugated to a horseradish for alkaloids. The compound was identified by its physical and spec-
peroxidase label. The liver specimens were finally treated with troscopic data; it was previously isolated from the same plant [48–50].
diaminobenzidine and then counterstained with hematoxylin. For PCNA To the best of our knowledge this is the first report on the complete
staining, a total of ten randomly chosen fields were evaluated and the assignment of the 1H and 13C NMR of tiliamosine (Supplementary data
results were expressed as the number of PCNA positive cells per 100 2).
hepatocytes.
3.2. In silico analysis of tiliamosine
2.3.5.8. Liver lipid quantification by sulphophosphovanillin In silico investigation of ADME properties of tiliamosine are given in
method. Quantification of liver lipids was done according to the supplementary Table 3 which showed tiliamosine lacked mutagenic,
method of Barnes and Blackstock [41]. One gram of liver sample was tumorigenic and irritant properties. Tiliamosine showed strong binding
homogenised in chloroform: methanol (2:1 ratio) mixture using affinity to the active site of FXRα at PRO`270/O, GLU`293/OE1/OE2
homogeniser thrice; the solvent portion was centrifuged at 3000 rpm (Fig. 2); WAY-362450 showed binding on FXRα at HIS`298/NE2
for 15 min; the supernatant was taken and made into 10 mL with (Fig. 3). The binding affinity and ligand efficiency of tiliamosine was
chloroform: methanol mixture. In a clean vial, 0.5 mL of the sample comparable with that of WAY-362450 (Table 1).
solution was taken, dried under nitrogen atmosphere, added with
0.5 mL of conc. H2SO4 and heated in a boiling water bath for 10 min. 3.3. Effect of tiliamosine on the viability of HepG2 Cells
The mixture was cooled; 5 mL of vanillin - phosphoric acid regent was
added with 0.2 mL acid digest, incubated for 30 min and the absorbance The effect of tiliamosine on the viability of HepG2 cells was ana-
was read at 530 nm. Olive oil was used as the standard and the results lysed using MTT assay and it was used for the fixation of doses for
were expressed as mg Tg/ g wet tissue. further in vitro studies. The treatment with tiliamosine did not exhibit
any toxicity up to 100 μM and it showed GI25 value of 264.28 μM. The
dose range of tiliamosine was fixed between 12.5 and 50 μM for in vitro
2.3.5.9. Preliminary assessment of mRNA expression of selected genes in studies (Fig. 4).
PO-BSA induced HepG2 cells and HFD-DEN induced rats. Primers of genes
such as PPARα, PPARγ, FXR, TNFα and β-actin were designed in 3.4. Effect of tiliamosine on lipid accumulation of PO-BSA induced steatotic
consultation with previously published literature [42–47]. The HepG2 cells
protocols and primer sequences are given in Supplementary data 3.
HepG2 cells incubated with PO-BSA complex showed 1.6 fold increment
in the ORO concentration; it indicated the increased accumulation of in-
2.3.6. Statistics tracellular Tg. The treatment with tiliamosine at all the selected con-
The values were given as mean ± SD for six animals. Statistical centrations brought down the ORO concentration. To confirm this finding,
significances of the data were assessed using one way analysis of var- intracellular Tg content was estimated; the reductions in intracellular Tg
iance followed by Tukey’s HSD (Graphpad Prism, Ver. 5.0, CA, USA) level at 25 and 50 μM concentrations were comparable with that of normal
and considered as significant at P ≤ 0.05. control and fenofibrate (Table 2). Microphotographs of HepG2 cells stained
968
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Fig. 7. Representative abdominal ultrasonogram of animals with HFD-DEN induced non-alcoholic steatohepatitis after four weeks of treatment with tiliamosine.
Scans were taken by B-mode ultrasound with Frequnce at 15 MHz and depth of 15 mm; (A) Normal control; (B) Negative control; (C) Sorafenib (60 mg/kg); (D)
Tiliamosine (50 mg/kg) and (E) Tiliamosine (100 mg/kg).
with ORO also substantiated the lipid lowering effect of tiliamosine in PO- also reduced the levels of transaminases in the spent media indicating
BSA induced steatosis (Fig. 5). the reduction of membrane damage. This was further confirmed by the
reductions in the MDA content in the treated cells (Table 2).
969
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
expressions of FXR; it lowered the expression of PPARγ (Supplementary tissue mass. Kidney and spleen showed statistically insignificant inter-
data 3). group variations (Table 3).
3.7. Acute toxicity study of tiliamosine 3.9. Effect of tiliamosine on the plasma biochemistry and liver lipid levels of
animals with HFD-DEN induced non-alcoholic steatohepatitis
Tiliamosine did not show any potential toxic effects on rats up to
2 g/kg concentration and it fell in the category 5 as per the OECD The treatment with tiliamosine significantly lowered the TC and Tg
guideline #423. The LD50 cut-off value of tiliamosine fell > 2 g/kg levels four weeks after treatment. The treatment did not alter the HDL-c
concentration and the dose range was fixed between 50–100 mg/kg. levels, but it significantly reduced the TC/HDL-c ratio. The liver lipid
levels were also significantly reduced by the treatment with tiliamosine
3.8. Effect of tiliamosine on relative organ masses in animals with HFD- (Table 4). The treatment with tiliamosine significantly altered the levels
DEN induced non- alcoholic steatohepatitis of phosphatases, transaminases, GGT and LDH levels; these reductions
were comparable with that of positive control (Table 5). In the treated
At the end of the treatment period, the disease control animals animals there was an improvement in the plasma albumin levels, but
showed 35.04% loss in the body mass compared to the normal control this increment was statistically insignificant (Table 6).
animals (Fig. 6). This can be correlated with the reduction in the feed
and water intake (Supplementary data 4). The treatment with tilia- 3.10. Effect of tiliamosine on the expression of selected genes in animals
mosine significantly improved the body weight and this increment was with HFD-DEN induced non-alcoholic steatohepatitis
comparable with that of normal control and sorafenib. The treatment
further lowered the relative liver mass and reteroperitoneal adipose As in the case of PO-BSA induced HepG2 cells, liver tissues from the
970
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
Fig. 9. Representative immunostained hepatic sections to show Proliferating Cell Nuclear Antigen (PCNA) expression from animals of HFD-DEN induced non-
alcoholic steatohepatitis after four weeks of treatment with tiliamosine.
Representative micrographs of (A) Normal control; (B) Disease control ; (C) Sorafenib (60 mg/kg); (D) Tiliamosine (50 mg/kg); (E) Tiliamosine (100 mg/kg) were
given. (F) Percentage of PCNA positive hepatocytes were counted from ten randomly selected fields at 400x magnification; Values (mean ± SD) having same
alphabet did not vary significantly (Tukey’s HSD; P ≤ 0.05).
treated animals showed the upregulation of FXR mRNA expression and mononuclear cell infiltration (Fig. 8). Hepatic sections were im-
downregulation in the mRNA expression of TNFα (Supplementary data munostained with PCNA to semi-quantitatively assess the hepatocyte
3). proliferation and this also showed that the treatment with tiliamosine
significantly reduced the number of PCNA positive cells (Fig. 9).
3.10.1. Ultrasound observation of effect of tiliamosine animal liver with
HFD-DEN induced non-alcoholic steatohepatitis 4. Discussion
Disease control animals showed mixed echogenicity and diffuse le-
sions in hepatic lobes; it indicated the presence of steatohepatitis. The NASH is one of the serious forms of NAFLD and its role on cirrhosis
animal treated with sorafenib and tiliamosine showed the homogenous and HCC was initially reported in 1990s; further studies demonstrated
echogenicity which showed the ameliorative effect of tiliamosine on that NASH as one of the risk factors of HCC [51]. So far, no specific
NASH (Fig. 7). pharmacological intervention was prescribed for the treatment of
NASH. NASH is connected with many pathways and molecules; thus
3.10.2. Effect of tiliamosine on the liver histology of animals with HFD-DEN there is a need to identify leads that target multiple pathways with
induced non- alcoholic steatohepatitis minimal adverse effects. Various natural products have been tried for
Administration of HFD and DEN to the animals induced congestion treatment of NAFLD; many phytochemicals were shown to have anti-
and hydropic degeneration of hepatocytes. The hepatocytes in the HFD- NAFLD efficacy by modulating various molecular pathways [52–54].
DEN control animals were non- eosinophilic and showed multifocal This study documented the curative effect of tiliamosine, a BBI alkaloid
mononuclear cell infiltration. The treatment with tiliamosine sig- on NASH.
nificantly improved the viability of hepatocytes and it reduced the In silico evaluation of the ADME properties of tiliamosine showed
971
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
972
S. Sylvester Darvin et al. Biomedicine & Pharmacotherapy 108 (2018) 963–973
[26] J. Dundas, Z. Ouyang, J. Tseng, A. Binkowski, Y. Turpaz, J. Liang, CASTp: computed (1990) 1020–1022.
atlas of surface topography of proteins with structural and topographical mapping [50] A. Ray, G. Mukhopadhyay, S. Mitra, K. Guha, B. Mukherjee, A. Nelofar,
of functionally annotated residues, Nucleic Acids Res. 34 (2006) W116–W118. Stereochemical aspects of diphenylbisbenzylisoquinoline alkaloids from Tiliacora
[27] M.F. Sanner, Python: a programming language for software integration and de- racemosa, Phytochemistry 29 (1990) 1023–1024.
velopment, J. Mol. Graph. Model. 17 (1999) 57–61. [51] L.A. Adams, J.F. Lymp, J.S. Sauver, S.O. Sanderson, K.D. Lindor, A. Feldstein,
[28] G.M. Morris, R. Huey, W. Lindstrom, M.F. Sanner, R.K. Belew, D.S. Goodsell, P. Angulo, The natural history of nonalcoholic fatty liver disease: a population-
A.J. Olson, AutoDock4 and AutoDockTools4: automated docking with selective based cohort study, Gastroenterology 129 (2005) 113–121.
receptor flexibility, J. Comput. Chem. 30 (2009) 2785–2791. [52] M. Zobeiri, T. Belwal, F. Parvizi, R. Naseri, M.H. Farzaei, S.F. Nabavi, A. Sureda,
[29] A. Stalin, S.S. Irudayaraj, G.R. Gandhi, K. Balakrishna, S. Ignacimuthu, N.A. Al- S.M. Nabavi, Naringenin and its nano-formulations for fatty liver: cellular modes of
Dhabi, Hypoglycemic activity of 6-bromoembelin and vilangin in high-fat diet fed- action and clinical perspective, Curr. Pharm. Biotechnol. 19 (2018) 196–205.
streptozotocin- induced type 2 diabetic rats and molecular docking studies, Life Sci. [53] M.H. Farzaei, M. Zobeiri, F. Parvizi, F.F. El-Senduny, I. Marmouzi, E. Coy-Barrera,
153 (2016) 100–117. R. Naseri, S.M. Nabavi, R. Rahimi, M. Abdollahi, Curcumin in liver diseases: a
[30] E. Toppo, S.S. Darvin, S. Esakkimuthu, A. Stalin, K. Balakrishna, K. Sivasankaran, systematic review of the cellular mechanisms of oxidative stress and clinical per-
P. Pandikumar, S. Ignacimuthu, N. Al-Dhabi, Antihyperlipidemic and hepatopro- spective, Nutrients 10 (2018) 855.
tective effects of Gardenin A in cellular and high fat diet fed rodent models, Chem. [54] I. Davoodi, R. Rahimi, M. Abdollahi, F. Farzaei, M.H. Farzaei, Z. Memariani,
Biol. Interact. 269 (2017) 9–17. F. Najafi, Promising effect of Rosa damascena extract on high-fat diet-induced
[31] R. Huey, G.M. Morris, A.J. Olson, D.S. Goodsell, A semiempirical free energy force nonalcoholic fatty liver, J. Tradit. Complement. Med. 7 (2017) 508–514.
field with charge‐based desolvation, J. Comput. Chem. 28 (2007) 1145–1152. [55] A. Moravcova, Z. Cervinkova, O. Kucera, V. Mezera, D. Rychtrmoc, H. Lotkova, The
[32] K. Stierand, M. Rarey, Drawing the PDB: protein− ligand complexes in two di- effect of oleic and palmitic acid on induction of steatosis and cytotoxicity on rat
mensions, ACS Med. Chem. Lett. 1 (2010) 540–545. hepatocytes in primary culture, J. Mod. Physiol. Res. 64 (2015) S627.
[33] E. Toppo, S.S. Darvin, S. Esakkimuthu, M.K. Nayak, K. Balakrishna, [56] C. Wang, Z.-L. Lv, Y.-J. Kang, T.-X. Xiang, P.-L. Wang, Z. Jiang, Aquaporin-9
K. Sivasankaran, P. Pandikumar, S. Ignacimuthu, N. Al-Dhabi, Effect of two an- downregulation prevents steatosis in oleic acid-induced non-alcoholic fatty liver
drographolide derivatives on cellular and rodent models of non-alcoholic fatty liver disease cell models, Int. J. Mol. Med. 32 (2013) 1159–1165.
disease, Biomed. Pharmacother. 95 (2017) 402–411. [57] E. Buzzetti, M. Pinzani, E.A. Tsochatzis, The multiple-hit pathogenesis of non-al-
[34] J. Ramirez-Zacarias, F. Castro-Munozledo, W. Kuri-Harcuch, Quantitation of adi- coholic fatty liver disease (NAFLD), Metabolism 65 (2016) 1038–1048.
pose conversion and triglycerides by staining intracytoplasmic lipids with Oil red O, [58] Y.-J. Chen, M.A. Wallig, E.H. Jeffery, Dietary broccoli lessens development of fatty
Histochemistry 97 (1992) 493–497. liver and liver Cancer in mice given diethylnitrosamine and fed a western or control
[35] S.S. Darvin, S. Esakkimuthu, E. Toppo, K. Balakrishna, M.G. Paulraj, P. Pandikumar, diet–3, J. Nutr. 146 (2016) 542–550.
S. Ignacimuthu, N. Al-Dhabi, Hepatoprotective effect of lawsone on rifampicin- [59] G. Yan, X. Wang, C. Sun, X. Zheng, H. Wei, Z. Tian, R. Sun, Chronic alcohol con-
isoniazid induced hepatotoxicity in in vitro and in vivo models, Environ. Toxicol. sumption promotes diethylnitrosamine-induced hepatocarcinogenesis via immune
Pharmacol. 61 (2018) 87–94. disturbances, Sci. Rep. 7 (2017) 2567.
[36] OECD, Test No. 423: Acute Oral Toxicity - Acute Toxic Class Method, OECD [60] M.A. Van Herck, L. Vonghia, S.M. Francque, Animal models of nonalcoholic fatty
Guidelines for the Testing of Chemicals, Section 4, OECD Publishing, Paris, 2002. liver disease—a starter’s guide, Nutrients 9 (2017) 1072.
[37] A.B. Nair, S. Jacob, A simple practice guide for dose conversion between animals [61] T. Inagaki, M. Choi, A. Moschetta, L. Peng, C.L. Cummins, J.G. McDonald, G. Luo,
and human, J. Basic Clin. Pharm. 7 (2016) 27. S.A. Jones, B. Goodwin, J.A. Richardson, Fibroblast growth factor 15 functions as
[38] P. Pandikumar, N.P. Babu, S. Ignacimuthu, Hypoglycemic and antihyperglycemic an enterohepatic signal to regulate bile acid homeostasis, Cell Metab. 2 (2005)
effect of Begonia malabarica Lam. in normal and streptozotocin induced diabetic 217–225.
rats, J. Ethnopharmacol. 124 (2009) 111–115. [62] T. Claudel, B. Staels, F. Kuipers, The Farnesoid X receptor: a molecular link between
[39] Y. Wang, L.M. Ausman, A.S. Greenberg, R.M. Russell, X.D. Wang, Nonalcoholic bile acid and lipid and glucose metabolism, Arterioscler. Thromb. Vasc. Biol. 25
steatohepatitis induced by a high‐fat diet promotes diethylnitrosamine‐initiated (2005) 2020–2030.
early hepatocarcinogenesis in rats, Int. J. Cancer 124 (2009) 540–546. [63] W. Huang, K. Ma, J. Zhang, M. Qatanani, J. Cuvillier, J. Liu, B. Dong, X. Huang,
[40] L.P. Van, Ketamine and xylazine for surgical anesthesia in rats, J. Am. Vet. Med. D.D. Moore, Nuclear receptor-dependent bile acid signaling is required for normal
Assoc. 171 (1977) 842–844. liver regeneration, Science 312 (2006) 233–236.
[41] H. Barnes, J. Blackstock, Estimation of lipids in marine animals and tissues: detailed [64] J. Li, R. Kuruba, A. Wilson, X. Gao, Y. Zhang, S. Li, Inhibition of endothelin-1-
investigation of the sulphophosphovanilun method for ‘total’lipids, J. Exp. Mar. Bio mediated contraction of hepatic stellate cells by FXR ligand, PLoS One 5 (2010)
Ecol. 12 (1973) 103–118. e13955.
[42] A.B. Schote, J.D. Turner, J. Schiltz, C.P. Muller, Nuclear receptors in human im- [65] S. Fiorucci, E. Antonelli, G. Rizzo, B. Renga, A. Mencarelli, L. Riccardi, S. Orlandi,
mune cells: expression and correlations, Mol. Immunol. 44 (2007) 1436–1445. R. Pellicciari, A. Morelli, The nuclear receptor SHP mediates inhibition of hepatic
[43] L. Zeng, W. Tang, J. Yin, L. Feng, Y. Li, X. Yao, B. Zhou, Alisol A 24-acetate prevents stellate cells by FXR and protects against liver fibrosis, Gastroenterology 127 (2004)
hepatic steatosis and metabolic disorders in HepG2 cells, Cell. Physiol. Biochem. 40 1497–1512.
(2016) 453–464. [66] Y. Liu, J. Binz, M.J. Numerick, S. Dennis, G. Luo, B. Desai, K.I. MacKenzie,
[44] Y. Xie, H. Wang, X. Cheng, Y. Wu, L. Cao, M. Wu, W. Xie, G. Wang, H. Hao, T.A. Mansfield, S.A. Kliewer, B. Goodwin, Hepatoprotection by the farnesoid X
Farnesoid X receptor activation promotes cell proliferation via PDK4-controlled receptor agonist GW4064 in rat models of intra-and extrahepatic cholestasis, J.
metabolic reprogramming, Sci. Rep. 6 (2016) 18751. Clin. Invest. 112 (2003) 1678–1687.
[45] F. Yan, Q. Wang, C. Xu, M. Cao, X. Zhou, T. Wang, C. Yu, F. Jing, W. Chen, L. Gao, [67] Y.D. Wang, W.D. Chen, M. Wang, D. Yu, B.M. Forman, W. Huang, Farnesoid X
Peroxisome proliferator-activated receptor α activation induces hepatic steatosis, receptor antagonizes nuclear factor κB in hepatic inflammatory response,
suggesting an adverse effect, PLoS One 9 (2014) e99245. Hepatology 48 (2008) 1632–1643.
[46] D.G. Remick, Applied molecular biology of sepsis, J. Crit. Care 10 (1995) 198–212. [68] R.M. Nijmeijer, R.M. Gadaleta, S.W. van Mil, A.A. van Bodegraven, J.B.A. Crusius,
[47] J.F. Antiabong, M.G. Ngoepe, A.S. Abechi, Semi-quantitative digital analysis of G. Dijkstra, D.W. Hommes, D.J. de Jong, P.C. Stokkers, H.W. Verspaget, Farnesoid X
polymerase chain reaction-electrophoresis gel: potential applications in low-income receptor (FXR) activation and FXR genetic variation in inflammatory bowel disease,
veterinary laboratories, Vet. World 9 (2016) 935. PLoS One 6 (2011) e23745.
[48] K. Guha, P. Das, B. Mukherjee, R. Mukherjee, G. Juneau, N. Bhacca, Structure of [69] X.-f. Huang, W.-y. Zhao, W.-d. Huang, FXR and liver carcinogenesis, Acta
tiliamosine: a new diphenyl bisbenzylisoquinoline alkaloid from Tiliacora race- Pharmacol. Sin. 36 (2015) 37.
mosa, Tetrahedron Lett. 17 (1976) 4241–4244. [70] Y.-D. Wang, W.-D. Chen, D.D. Moore, W. Huang, FXR: a metabolic regulator and
[49] A. Ray, G. Mukhopadhyay, S. Mitra, K. Guha, B. Mukherjee, A. Nelofar, A diphe- cell protector, Cell Res. 18 (2008) 1087.
nylbisbenzylisoquinoline alkaloid from Tiliacora racemosa, Phytochemistry 29
973