Fisiopatologia Do Ferro Na Neurodegeneração - 2019
Fisiopatologia Do Ferro Na Neurodegeneração - 2019
Brain Iron
Metabolism and
CNS Diseases
Advances in Experimental Medicine and Biology
Volume 1173
Editorial Board
Irun R. Cohen, The Weizmann Institute of Science, Rehovot, Israel
Abel Lajtha, N.S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
John D. Lambris, University of Pennsylvania, Philadelphia, PA, USA
Rodolfo Paoletti, University of Milan, Milan, Italy
Nima Rezaei, Children’s Medical Center Hospital, Tehran University of Medical
Sciences, Tehran, Iran
Advances in Experimental Medicine and Biology presents multidisciplinary and
dynamic findings in the broad fields of experimental medicine and biology. The
wide variety in topics it presents offers readers multiple perspectives on a variety of
disciplines including neuroscience, microbiology, immunology, biochemistry,
biomedical engineering and cancer research.
Advances in Experimental Medicine and Biology has been publishing excep-
tional works in the field for over 30 years and is indexed in Medline, Scopus,
EMBASE, BIOSIS, Biological Abstracts, CSA, Biological Sciences and Living
Resources (ASFA-1), and Biological Sciences. The series also provides scientists
with up to date information on emerging topics and techniques. 2018 Impact Factor:
2.126
123
Editor
Yan-Zhong Chang
College of Life Sciences
Hebei Normal University
Shijiazhuang, Hebei, China
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Contents
v
Contributors
vii
viii Contributors
Abstract Iron is the most abundant trace element in the human body. It is well
known that iron is an important component of hemoglobin involved in the transport
of oxygen. As a component of various enzymes, it participates in the tricarboxylic
acid cycle and oxidative phosphorylation. Iron in the nervous system is also involved
in the metabolism of catecholamine neurotransmitters and is involved in the forma-
tion of myelin. Therefore, iron metabolism needs to be strictly regulated. Previous
studies have shown that iron deficiency in the brain during infants and young chil-
dren causes mental retardation, such as delayed development of language and body
balance, and psychomotor disorders. However, if the iron is excessively deposited
in the aged brain, it is closely related to the occurrence of various neurodegenerative
diseases, such as Alzheimer’s disease, Parkinson’s disease, and Friedreich’s ataxia.
Therefore, it is important to fully study and understand the mechanism of brain iron
metabolism and its regulation. On this basis, exploring the relationship between brain
iron regulation and the occurrence of nervous system diseases and discovering new
therapeutic targets related to iron metabolism have important significance for break-
ing through the limitation of prevention and treatment of nervous system diseases.
This review discusses the complete research history of iron and its significant role in
the pathogenesis of the central nervous system (CNS) diseases.
Iron is an essential player of many biological processes in the brain such as DNA
synthesis, oxygen transport, myelin formation, and mitochondrial functions. It is
necessary to maintain the iron homeostasis for normal physiological actions of the
After the bronze era, Iron Age (~1200–1000 BC) began to dominate every creature
in the universe, and it turned into an imperative player of entangled biochemistry,
which means iron is indispensable for life. Iron is one of the most superabundant
metals that contribute to make earth habitable by generating magnetosphere circling
to the planet. Indeed, oxygen-free states of the earth made possible to occur a num-
ber of chemical reactions that initiate the life existence. Importantly, the essential
compound that possibly creates life is a soluble form of iron, hydrogen, and sul-
fide from ores. In the earlier life formation, the soluble iron was rich, but it began
to reduce due to oxygen addition that means life formation is affected slowly but
steadily. Günter Wächtershäuser proposed that chemistry of life occurred in the min-
eral sources [60, 61]. In the sixteenth century, Nicholas Monarde used the iron for
various ailments such as alopecia, acne, wounds, hemorrhoids, and gout, revealed
the biological significance of iron [42].
The underlying biological importance was proposed initially by Lemmery and
Geofgroy in 1713 [57]. Nonetheless, it was nearly required two centuries to compre-
hend iron metabolism in the body. Vincenzo Menghini was discovered the presence
of iron in the blood at the earlier of seventeenth century in the human body [4]. This
group was scorched the blood from various models including birds and humans for
searching iron in the blood. In the eighteenth century, Louis Rene Lecanu discovered
that RBC has a protein called globulin and red coloring matter called hematosine
that contained iron [34, 35]. In the earlier nineteenth century, Fontés and Thivolle
recommended an imperative revelation that a body has unique form of iron in rela-
tion to the hemoglobin in the serum [18]. They additionally found that inadequacy of
iron can affect the serum iron concentration. In the mid of nineteenth century, Holm-
berg and Laurell found the iron-binding proteins called transferrin (Tf) [26]. In the
meantime, Schade and Caroline determined the antibacterial effect of iron-binding
proteins, and they proposed that siderophilin, a bacterial iron-absorbing compound,
1 Brain Iron Metabolism and CNS Diseases 3
which was indistinguishable to Tf proteins [52]. This finding has crucial informa-
tion since most of the drugs that are currently used for chelation therapy belong to
siderophilins. Henry John Horstman Fenton proposed that iron can become toxic in
certain circumstances when the level of iron is surpassing in the cell [14, 15], and the
concept was not acknowledged by the researchers until the disclosure of superoxide
dismutase which converts superoxide into oxygen and hydrogen peroxide.
Studies from 1937 by John Livingood, Fred Fairbrother, and Glenn Seaborg found
an exceptional radioactive labeled iron for analyzing iron homeostasis. Based on
this radioactive iron, McCance and Widdowson in 1938 proposed that iron is not
excreted from the body [44]. Likewise, they theorized that only a small amount of
iron was assimilated from the whole intestinal tract, but the main site of assimilation
occurred at the upper locale of the intestine. Their investigation was accurate with
respect to iron homeostasis except their suggestion that “in hemochromatosis, the
iron deposited in the cells is insoluble and inert, and takes no part in the general
equilibrium. Hence there is a true deficiency of free iron, as evidenced first, by the
anemia, and secondly, by the cure of the anemia by the administration of iron and
for their estimation that the red cell life span was only 30 days.” Ferrokinetic studies
by Pollycove and Mortimer in 1961 were proposed a clear and surmised size of each
iron pool [49].
In 1997, Hediger’s group and Andrews’ group discovered an important protein
called divalent metal protein 1 (DMT1) also known as DCT1 and Nramp2 which
transports the dietary iron using H+ electrochemical gradient as driving force. They
found that the expression of DMT1 in the rat increased the uptake of 55 Fe2+ and Fe2+ .
They also found that the DMT1 expression was reactive to several other divalent metal
ions. Vulpe group discovered a multicopper oxidase protein called hephaestin in the
small intestine in 1999 and Chen group in 2010 identified another type of multicop-
per ferroxidase called zyklopen in the placenta, and the expression of these proteins
is necessary for iron efflux from placental cells and small intestine [6, 59]. The
iron entry into plasma, iron utilization and storage are determined by the interaction
of the peptide hormone called hepcidin and ferroportin (FPN1). FPN1 was identi-
fied by three different groups using different methods. The first group found FPN1
through zebrafish mutant gene [11]. The second group identified FPN1 as a transcript
[39]. This group also identified the Dcytb (duodenal cytochrome b) (also known as
Cybrid1) as an iron-regulated protein using a subtractive cloning strategy [40], and
the third group identified the FPN1 as an iron-responsive element (IRE)-containing
mRNA [55]. The role of hepcidin in relation to iron regulation was identified by Nico-
las group in 2001 through the HAMP1 gene deletion, which encodes hepcidin [45].
All of these discoveries have been greatly promoted to understand iron metabolism
in the body.
4 A. Thirupathi and Y.-Z. Chang
Understanding of iron metabolism begins with dietary iron that can be efficiently
taken to the cells through duodenal enterocytes. This process is firmly managed
by several sensory on and off mechanisms in the epithelium, which is carried out
by a number of proteins such as DMT1, cytochrome b ferrireductase, ferroportin,
TfR1 and hephaestin. Several enigmatic concepts existed in earlier studies about iron
absorption. One of the concepts proposed by Hahn and Whipple is known as “mucosal
block mechanism.” They declared that the first dose of given iron is saturated the
mucosa with ferritin for blocking the absorption of the second dose [21]. In the
1950s and 1960s, some studies had contended against mucosal block mechanism that
anemia can be upregulated by the iron absorption in the iron deficiency condition,
and it was reduced by iron overloading condition. Although studies had given a clear
idea about iron absorption by bowel in the middle of the twentieth century, the bowel
components that direct iron absorption are still elusive. Conrad and Crosby in 1963
demonstrated that the entry of radioactive iron in mucosa sloughed off when the
body needs low iron [8, 9]. They inferred that the iron regulation was determined
by the exchange of iron between mucosal to blood, and the concept is now widely
accepted. However, the mechanism remains elusive.
The intestinal mucosa responds if the body’s iron level is altered and facilitates the
iron absorption accordingly. Iron deficiency condition increases absorption, whereas
iron overload condition decreases the iron absorption. The loss of iron occurs through
desquamation process and an adult man losses 1 mg of iron, and this level varies with
women during their menstruation periods. The healthy population needs 1–2 mg of
iron per day for compensating the loss of iron. Dietary iron absorption takes place
in the duodenum and upper jejunum. Several proteins are involved to carry out this
process such as DMT1, FPN1, and Dcytb. The dietary irons are mostly as inorganic
form, and it is reduced by the Dcytb, a ferrireductase enzyme present in the brush
border of microvilli. Then, DMT1 transports the ferrous form of iron from the api-
cal membrane of the duodenal enterocytes into intestinal epithelial cells where it
can be stored as ferritin or transported into circulation across the basolateral mem-
brane. FPN1 transports the iron from the basolateral membrane to circulation and
non-transported iron is removed from the body through exfoliation of the intesti-
nal epithelium. This epithelium exfoliation may represent the iron excretion because
these cells are expressing TfR1 at the basolateral membrane and iron from the plasma
can enter through endocytosis. For transporting iron through the basolateral mem-
brane, iron must be transported through cell cytoplasm, but this mechanism is not
fully understood. However, this iron transport possibly occurs through cytosolic
chaperons and iron-binding proteins or transcytosis. Iron transport from the basolat-
eral membrane of enterocyte to circulation requires the changes of redox state which
is carried by the hephaestin in the duodenum and ceruloplasmin (CP) in other parts
1 Brain Iron Metabolism and CNS Diseases 5
of the body which converts the intracellular Fe(II) back to extracellular Fe(III) for
transferrin [59].
Several researchers are involved to explore the cellular level of iron homeostasis.
We referenced here some of the remarkable researches regarding iron homeostasis.
Finch group first investigated the reticulocytes iron transference in 1949 [17]. In
little later, Jandl and Katz groups proposed the presence of TfR1 for take-up irons
from the reticulocytes [30]. Evan Morgan and his group were evidenced by the cel-
lular level of iron accession mechanism of Tf in the 1970s [2, 43]. Furthermore,
Fineberg and Greenberg provided the molecular level of iron accession mechanism
[16]. They proved that the administration of iron increases the ferritin protein syn-
thesis, and this experiment was further carried out by Drysdale and Munro [12, 63].
Zahringer group detailed that iron administration increased the ferritin mRNA con-
tent in the actinomycin-treated animals. Munro group explained that iron-sensitive
factors called iron-responsive elements (IREs) and iron regulatory proteins (IRPs)
can regulate ferritin mRNA translation. They reported that ferritin availability to iron
was decreased by 5 UTR deletion containing a conserved 28 base sequences from
ferritin mRNA [33]. Some earlier studies reported that TfR1 and ferritin expression
are absolutely inverse. When iron is overabundance, the level of TfR1 is constrained;
whereas the iron is less, the level of TfR1 is high. Therefore, ferritin and TfR1 levels
are precisely altered at the cellular level. Owen and Kuhn evidenced that the sequence
within the 3 UTRs is needed for iron-dependent TfR1 expression, while TfR1’s pro-
moter region is not needed [47]. Further research evidenced that IREs present in
ferritin while TfR1s bind with an IRE-binding proteins called IRPs. There are two
types of IRPs having almost similar function. Higher iron can increase the translation
of ferritin mRNA and decrease the stability of TfR1 mRNA. This crucial mechanism
is orchestrated by the presence of IREs at the 5 UTR of ferritin mRNA and 3 UTR
of the TfR1 mRNA. These IREs of both ferritin and TfR1 can bind with cytosolic
proteins IRP1 and IRP2. Cells acquire iron from the plasma Tf. After loading iron to
Tf, the complex can bind with TfR1 on the cell surface and undergoes endocytosis
through clathrin-coated pits. For triggering the release of iron from the Tf, proton
pump induces the acidification of endosome to pH level 5.5. For redox changing to
transferring iron by DMT1, ferrireductase STEAP3 reduces Fe3+ to Fe2+ , and this
Fe2+ form can be stored in ferritin or utilized by downstream metabolic pathways.
After releasing iron from Tf, the Tf is dissociated from the TfR1 and the apo-Tf is
entered into circulation where it can recapture the Fe3 . Regulation of cellular iron
homeostasis is tightly managed by the post-transcriptional mechanism, and especially
the IRP-IRE system can mediate this post-translational modification for influencing
the synthesis of proteins that are responsible for maintaining the intracellular iron
level. In the iron depletion condition of a cell, IRPs bind with IREs in the 5 UTR or
3 UTR of mRNA for either suppressing the translation of mRNA such as FTH1 and
6 A. Thirupathi and Y.-Z. Chang
FPN1 or by increasing the mRNA translation including TfR1 and DMT1 [24, 25].
Under cellular iron increased conditions, IRPs loses their binding activity to IREs
by acquiring iron–sulfur cluster (4Fe–4S cluster) and proteasomal degradation [20,
51].
In the brain, the function of iron in DNA synthesis, gene expression, neurotransmis-
sion, myelination of neurons, and the mitochondrial system can be crucial. There-
fore, the balance of brain iron needs to be strictly regulated. However, iron abnormal
metabolism can lead to misbehave many proteins that are associated with brain
diseases or disorders. The higher iron deposition or iron deficiency may impair the
normal functions of the cells. Over iron accumulation in a specific region of the brain
leads to several neurodegenerative diseases such as Alzheimer’s disease and Parkin-
son’s disease. Dysregulation of iron homeostasis also sets diseases like Friedreich’s
ataxia (FA). Understanding iron metabolism in the CNS is increasing interest. In
addition, numbers of questions are travelling along with scientific discoveries. For
example, what are the characteristics of iron metabolism and its regulatory mecha-
nisms in each cell type of the CNS such as neurons, oligodendrocytes, and microglia
and what is the mechanism of iron transport and regulation across the blood–brain
barrier?
The brain requires iron for its metabolic process like other organs. However, each
type of brain cells such as neurons, oligodendrocytes, and astrocytes possess distinct
iron metabolism. In addition, unequivocal concepts exist that how iron cross the
blood–brain barrier (BBB) or cerebrospinal fluid (CSF) from the circulation and
how it traffics in the brain parenchyma to provide iron to different brain cells. The
distribution of iron in the brain is mostly concentrated in the substantia nigra pars
compacta (SN), circumventricular organs, globus pallidus, and oligodendrocytes. It
is well known that the brain is protected by a special structure called BBB, which
can prevent the pathogen and other macromolecular compounds entry into the brain
and ventricles. Therefore, transporters and receptors can mediate iron transfer across
the luminal membrane of endothelial cells of BBB into the CNS. Nevertheless, these
regulatory molecules are associated with many diseases and are the major baffling
of current research.
The current understanding of iron homeostasis in the brain begins with the Tf/TfR
pathway, may be the major route for iron transport across the luminal membrane of
the endothelium. Transferrin-bound iron then be converted into Fe3+ and translo-
cated across the endosomal membrane by DMT1. Tf will be returned to the luminal
1 Brain Iron Metabolism and CNS Diseases 7
membrane once it releases iron with TfR. Other pathways may also be involved
to transport iron across the BBB such as lactoferrin receptor/lactoferrin and GPI-
anchored melanotransferrin/soluble melanotransferrin. In addition to these pathways,
transferrin-bound iron is transferred across BBB through transcytosis. FPN1 and hep-
haestin might play a key role in transferring iron across the BBB as Fe2+ . Another
possible way for iron transfer for across the BBB is carried out by astrocytes. The
FPN1 released Fe2+ in basolateral surface of endothelial cells. Then, astrocytes oxi-
dize Fe2+ into Fe3+ by CP through their end-feet processes on the capillary endothelia
[38, 46], which are further bound with Tf produced by choroid plexus and oligoden-
drocytes, suggesting that iron not only enters via endothelial cells of BBB but also
enters through epithelial cells of choroid plexus. After entering iron into interstitial
fluid or CSF in ventricles, the iron will binds with Tf and diffuses via CSF and
interstitial fluid of the brain parenchyma, supply iron to the cells expressing TfR
within the CNS. A small amount of transported iron can circulate with citrate and
ATP. Different cell types in the brain are involved to acquire iron using different
pathways. Neurons acquire iron through Tf, astrocytes acquire iron through DMT1,
oligodendrocyte acquires iron from Tf by the TfR1 pathway and from ferritin via
TIM-2 receptors, and microglia acquire iron from ferritin. Although several studies
are well established the iron homeostasis in the CNS, due to BBB, the iron levels in
the peripheral nervous system (PNS) decouple from the brain. However, FPN1 may
play the role to export the iron from the CNS to PNS with the help of ferroxidase.
Furthermore, PNS has little information regarding iron homeostasis. Since BBB has
limited access to iron, it expresses all the iron regulatory proteins as we mentioned
above. However, on the contrary, the peripheral nervous system has little informa-
tion regarding iron regulatory proteins. The individual nerve fiber is protected by
the connective tissue matrix like epineurium, perineurium, and endoneurium in the
PNS. Unlike BBB, the blood–nerve barrier (BNB) permits the proteins and macro-
molecules. Both BBB and BNB shared the common feature in transferring iron, such
as having enzymes, receptors, and transporters. For example, Tf-bound iron is the
main source for CNS, but other sources can also contribute to transfer iron in the
CNS. However, PNS depends on Tf–TfR1 pathway for iron acquisition especially Tf
is accumulated in the Schwann cell (SC) cytoplasm, suggesting a role of Tf-bound
iron in the myelination. Furthermore, SC expresses other iron exporters like FPN1
and ferroxidase CP which associate together to efflux the iron to the axons in the
peripheral nerve. CNS has ferritin as an iron source, but PNS does not have a ferritin-
dependent active mechanism [23]. PNS acquires iron using NTBI through DMT1
[58].
Iron passage into the brain had been a long mystery. Initially, it was believed that
the iron entry occurred before the blood–brain barrier maturation at the infant stage.
Now, it is known that endothelial TfR1 mediates the brain iron uptake. However, TfR
8 A. Thirupathi and Y.-Z. Chang
Fig. 1.1 Understanding of iron homeostasis in the brain. TfR1 of endothelial junctional complexes
and Tf can transfer the iron with the endosomal process. Detachment of iron from Tf, can be pumped
into the cytosol by DMT1 where it can be stored as soluble form of iron. Some iron can be pumped
out by FPN1, which is further oxidized by ceruloplasmin into ferric form for Tf. Non-bound iron
can be either attached with ATP or citrate which is released from astrocytes
Although brain cells have tight regulation for iron entry, iron is the primary source
for several cytosolic proteins and enzymes, mitochondria, and nuclear ferroproteins
of brain cells. It has been known that iron accumulation is directly proportional
to aging. However, this situation is not connected with pathogenesis. Some special
regions such as globus pallidus and substantia nigra in the brain can participate to
increase the iron deposition, but factors causing iron deposition in those regions are
unknown. Possibly, the aging condition can synthesize some special type of neu-
rons to increase the expression of iron-storing protein ferritin. For example, globus
pallidus has rich iron level as equivalent as in the hepatocytes. Different scavenger
cells including macrophages can become iron-rich when the brain cells started to
undergo apoptosis. IRP1 and IRP2 may also play important roles in controlling the
cellular iron homeostasis in the brain. Alterations in these IRPs signaling pathways
can contribute to higher iron deposition in the brain. For example, iron deficiency
condition can allow IRP1 to interact IREs for up-regulating the TfR expression in
10 A. Thirupathi and Y.-Z. Chang
the brain endothelial cells, but alterations in these signaling pathways can increase
the brain iron accumulation.
CSF is the major route for iron export from the brain into the blood from the sub-
arachnoid space. Interference in the CSF route can cause the over iron accumulation
in the brain. For example, transferrin is very low in the CSF, thus exporting iron
from the brain to blood is limited. Some other iron-storing proteins like lactoferrin,
ferritin, and non-protein-bound iron can help to export the iron, but these proteins
function is majorly affected by some pathological conditions. In addition, microglia
and other phagocytic cells are involved to export the iron. Mitochondrial proteins
can also involve in exporting iron. For example, iron/sulfur (Fe/S) clusters are acted
as a prosthetic group of respiratory complexes which can contribute to eliminate the
iron from mitochondria. In other way, the storage protein ferritin can reduce the free
from iron in the cytosol by sequestering them, but changes in this export mechanism
can contribute to increase iron accumulation in the brain. In a simple description,
age factors, genetic mutation, and inflammation can cause the iron accumulation in
the brain [56, 62].
Iron deposition in a particular region of the brain can mishandle the iron metabolism
because this region is already rich with iron, and thereby causing various distinct
disorders called neurodegeneration with brain iron accumulation (NBIA). Neurode-
generation with brain iron accumulation is mainly linked with the mutations in the
iron homeostasis genes and is characterized by gait problems, movement dysfunction,
and motor–cognitive problems. Until now, there are fifteen major genetically distinct
disorders which can be discussed in further content. However, other neurodegener-
ative diseases like Parkinson’s disease, Alzheimer’s disease, and Hunting’s disease
can also be believed to be caused by secondary iron accumulation. This is not nec-
essarily the case. It may also be a disease caused by existing iron deposits (such as
age and inflammation).
Parkinson’s disease (PD) is one of the most extensive diseases that relate to dys-
regulation of iron metabolism. Although the causes of PD have been very clear, the
initial discovery of PD was written in 1817 by Parkinson [48]. After this, Jean-Martin
Charcot expanded this research internationally [5]. Since then, several concepts are
associated with PD causes. Recent research has received immense interest to find the
role of iron in causing PD. Patients with PD have higher iron accumulation in the
pars compacta area of the substantia nigra. Several MRI studies have also observed
that elevated level of iron is associated with PD. The cause of PD is linked with the
1 Brain Iron Metabolism and CNS Diseases 11
Abnormal iron metabolism has been considered as a vital marker for the earlier
setting of Alzheimer’s disease (AD). However, the link of iron accumulation to the
AD pathology is not well established. Studies have observed that the patients with
AD have elevated level of iron in the cortical, subcortical, and white matters of the
brain. Amyloid (Aβ) plaque aggregation in a specific region of the brain has been
considered as AD pathology, and Aβ is formed by proteolytic cleavage of amyloid
precursor protein (APP). APP is cleaved by α-secretase and β-secretase. Normally,
alpha pathway is not involved in the generation of amyloidogenesis, whereas beta
pathway is involved in the amyloidogenesis. Overproduction of Aβ42 and alloforms
of Aβ can lead the AD pathogenesis, and the connection of APP and Aβ alloforms
with abnormal iron metabolism has been observed in AD patients. Both ferrous and
ferric forms of iron can interact with the Aβ alloforms. However, Aβ42 fragments
can form amyloid more rapidly. Aggregation of Aβ42 can form monomer, dimers,
oligomers, fibrils, and senile fibril plaques. The ferric form of iron can bind with all
forms of Aβ42. However, it has a difficulty to break from the fibrils, but this plaque
formation with ferric form can initially remove the excess-free form of iron in the
brain. However, the ferrous form can also bind with Aβ42 to revert the function
of amyloid formations. The core of matured amyloid plaque has both ferrous and
ferric form of iron, and the core of amyloid plaque is the place of oxidative stress.
In addition, the ferroxidase activity of APP can prevent the FPN1-mediated iron
loading to Tf resulting in iron retention in the neurons and further causing neuronal
death. Alzheimer’s disease brains have increased the accumulation of iron in the
specific region of the brain such as the hippocampus and cerebral cortex. Aβ plaques
and neurofibrillary tangles have reactive iron and are the important site for catalytic
12 A. Thirupathi and Y.-Z. Chang
Genetic analysis of iron overload condition has revealed a number of target proteins
that control iron homeostasis, and the mutation in these proteins disrupts the iron
homeostasis. Molecular level of research has identified the several loops that control
the iron homeostasis and is mounted in the hepcidin–ferroportin interaction. This
regulatory pathway accounts genetic mutation of iron overloading condition and
related disorder. PKAN is a neuronal iron overloading disease linked with the gene
encoding pantothenate kinase 2 (PANK2) mutations, a crucial enzyme present in
the mitochondrial inter-membrane space and synthesis mitochondrial coenzyme A
from pantothenate. PKAN is previously known as Hallervorden–Spatz syndrome.
This disease is characterized by neurological impairments such as childhood onset
dystonia and spasticity. Study has demonstrated that lack of Pank2−/− in the mice
did not have iron overload causing neurological symptoms in the CNS [64]. Patients
with PKAN have reduced cholesterol level and fatty acid synthesis, and also the
Pank2−/− mice have reduced mitochondrial potential and ATP generation. However,
the exact reason of iron overload in the brain regions is not entrenched, particularly,
why PANK2 associated iron overload in the globus pallidus, the possible reason is
that the other homologues such as PANK1 and PANK3 may be too low to compensate
the loss of PANK2. Likewise, the neurons can cause the iron overload before they
die, which results in iron overload neuron loss in PKAN.
PLAN is linked with different mutations in the PLA2G6 gene, and the symptoms
are dystonia, spasticity, and cerebellar atrophy. PLA2G6 gene encodes phospholipase
A2 group VI, a critical regulator of mitochondrial membrane stability and remodel-
ing. MRI result has shown that iron deposition presents in the globus pallidus and
1 Brain Iron Metabolism and CNS Diseases 13
substantia nigra and striatum varies. PLA2G6 gene producing enzymes regulate fatty
acid synthesis in the brain. However, it is believed that acetyl CoA can recompense
the needs of fatty acid in the brain. MPAN is a condition associated with childhood
or early adulthood. MPAN affected people can have movement problems including
spasticity and dystonia. Other neurological issues such as degeneration in the nerves
that carry information to the brain (optic atrophy), dysphagia and incontinence are
linked with MPAN. Mutations in the C19orf12 can cause MPAN. C19orf12 encodes
a mitochondrial membrane protein which can participate in the fatty acid metabolism
and branched-chain amino acids. MPAN is characterized by iron over accumulation
in the globus pallidus and substantia nigra. In MPAN, Lewy bodies containing abnor-
mal alpha-synuclein present throughout in the globus pallidus, deep gray matter, and
neocortex.
Aceruloplasminemia is characterized by the mutation of CP. It was initially
described in 1987. Ceruloplasmin is a multicopper ferroxidase, lost its ferroxidase
activity in aceruloplasminemia condition. As a result, CP may not be able to convert
the ferrous iron form into ferric form to transferrin, which results to fail the export
iron out of brain astrocytes and macrophages to the circulatory system. Patients with
aceruloplasminemia can have higher iron overload in the astrocytes, and these regions
of the brain lose the neurons, which result in the triad of adult-onset neurological
disease and retinal degeneration. The deformity of astrocytes is a prominent charac-
teristic of aceruloplasminemia brains. The proportion of iron deposition is correlated
with the globular structure of the astrocytes [41]. In addition, the iron does not reach
to the neurons due to inactivity of CP, and thus, neurons are susceptible to die.
Neuroferritinopathy is an autosomal dominant neurodegenerative disease caused
by ferritin light chain 1 (FTL1) light chain mutations. Curtis and groups identified
first time neuroferritinopathy in 2001 [7]. Until now, there have been ten mutations
reported to cause this disease. Additional nucleotide inclusion due to a mutation in
the FTL1 can neglect to sequester the iron, and consequently iron leaks out of the
ferritin. This condition makes the cytosol to turn out to be progressively oxidized
environment. Ferritin can also respond to this oxidized cytosolic environment, and
this can further allow more leakage of iron. Furthermore, higher accumulation of iron
can make the IRE-binding proteins to not turn IRE-binding form which further results
in the overproduction of ferritin and ferritin aggregation with iron in the neurons,
substantia nigra, glial cells of the globus pallidus, and cerebellum. Initial failure
of iron sequestration can lead the higher transcription, and reduced translational
repression in the FTL can allow this abnormal protein segregation in the various
regions of the brain. FAHN is caused by the mutation in the gene encoding fatty acid-
2-hydroxylase gene (FA2H) and is involved to produce fatty acid for sphingolipids.
Defect in this gene is associated with abnormal myelination. Studies have shown that
FAHN patients have iron accumulation in the globus pallidus, substantia nigra, and
subcortical and periventricular regions. Also, ferritin-loaded microglial cells enter
into the areas of regenerating tissue for further iron loading. FAHN is characterized
by the spasticity, ataxia/dystonia, and optic atrophy.
14 A. Thirupathi and Y.-Z. Chang
Several serendipitous findings are observed for treating neurological diseases because
these conditions are different from other disorders, and the evaluation therapy has
been designed based on the developing information of biochemistry, physiology,
and anatomy of the brain. For example, Parkinson’s disease is one of the impor-
tant neurological conditions, and treatment for this disease is tightly based on the
above-mentioned merit. Parkinson was believed that the “progress of the disease may
be stopped” and he deliberately inserted a small piece of cork into the blisters and
inflammatory skin for expelling the pressure away from the brain and spinal cord.
Charcot and groups were first established the treatment for PD using belladonna alka-
loids. These alkaloids influence the cholinergic/dopaminergic balance in the striatum
for improving PD. Since then, many active anticholinergic agents have been devel-
oped for treating PD. However, Charcot wanted to use hyoscyamine. He occasionally
used hyoscyamine with rye-based ergot products, a pharmacological dopamine ago-
nist used to invigorate the striatal dopamine receptors. He inferred in 1872 that he
attempted almost everything to treat PD, but all of them were rejected. Then, Char-
cot tried vibratory therapy for managing PD, and he found that patients with PD had
decreased symptoms. However, the tremor was not improved. He used a combina-
tion of hyoscyamine with arsenic, morphia and cannabis, and opium and he observed
that the patients with PD have distinct improvements [19]. The modern therapy is
used cannabis for dopaminergic activation. Although several anticholinergic drugs
were developed in the nineteenth and twentieth centuries, all of them were similar
effects. Holtz discovered an important enzyme called dopa carboxylase that can con-
vert levodopa to dopamine [29]. Loss of dopamine leads the pathogenesis of PD.
Ehringer and Horneykiewicz observed that the depletion of dopamine in the human
brains [13, 28]. Birkmayer and Horneykiewicz injected the levodopa intravenously
first time to the PD patients, and they observed significant improvement in the PD
[3]. For searching levodopa in the naturally available plants, Manyam observed that
cowitch plant (Mucuna pruriens) contains levodopa [37]. It is pointed out that the
current treatment of PD defects, especially the late use of L-DOPA, has no effect,
and the understanding of iron metabolism is helpful for the treatment of PD.
AD is one of the most leading dementia diseases. Although the number of scien-
tific effort has been made to prevent the disease, there are no effective pharma-
cotherapeutic options available for treating AD. To date, several drugs have been
prescribed based on symptomatic approach for counterbalance the neurotransmitter
1 Brain Iron Metabolism and CNS Diseases 15
disturbances in the AD. Cholinergic hypothesis has been linked with AD including
loss of acetylcholine neurons and acetylcholine synthesis and degradation. There
are three approved drugs named cholinesterase inhibitors (CI) used for enhancing
the cholinergic transmission like donepezil, rivastigmine, and galantamine. These
CIs have been shown to improve the cognitive functions of AD. Another therapeutic
option for AD is memantine, and this drug can bind with N-methyl-D-aspartate antag-
onist for protecting neurons. In addition, a combination of donepezil with memantine
significantly improved the cognitive function of moderate to severe AD patients [27,
54]. Another cause for AD is amyloid (Aβ) plaque aggregation. Several researches
have been driven to finding a compound for preventing Aβ aggregation. The only Aβ
aggregation inhibitor is a synthetic glycosaminoglycan 3-amino-1-propanesulfonic
acid (3APS, tramiprosate). Tramiprosate interferes with the Aβ aggregation. How-
ever, data suggest that tramiprosate increases the abnormal aggregation of Aβ [53].
Other compounds such as Colostrinin and scyllo-Inositol can interfere with the Aβ
aggregation. However, the specificity of those compounds is questionable. Drugs
such as PBT2 is a second-generation drug of 8-OH quinoline interferes with the
Cu2+ and Zn2+ mediated toxic oligomerization of Aβ. Also, several compounds that
inhibit BACE (β-site AP-cleaving enzyme inhibition) and γ-secretase are in the pre-
clinical stage. Therefore, the current clinical trials for the treatment of AD drugs have
failed because the mechanism of AD pathogenesis has not been fully understood,
so the understanding of iron metabolism may contribute to the development of AD
drugs.
Therapies for NBIA disorders are symptomatic. NBIA is associated with different
genes mutations that are linked with iron metabolism-regulating proteins. Therefore,
understanding a common feature that shares to induce the pathogenesis in NBIA can
help to develop therapies for NBIA disorders. For example, aceruloplasminemia and
neuroferritinopathy have common defects in handling iron, which results to increase
the oxidative damages, whereas other NBIA disorders such as PLAN and MPAN
are linked with lipid metabolism and mitochondria. Therefore, finding a common
link with membrane homeostasis, mitochondrial function, and iron metabolism may
advantage therapeutic development. Oxidative stress is one of the common condi-
tions of above-mentioned situations. Therefore, understanding of the mechanism
that induces iron metabolism causing oxidative stress and its further consequences
in developing NBIA may help to design a therapy for NBIA-associated disorders.
16 A. Thirupathi and Y.-Z. Chang
1.6 Conclusion
The present drugs for treating iron overload causing neurodegeneration are symp-
tomatic and do not able to prevent the progression of the diseases. Nevertheless, these
drugs are giving consistent improvements for cognition and global status. In addition,
the search for disease-modifying approach for treating these diseases is unsuccess-
ful in demonstrating the efficacy in the clinical stages. Meanwhile, the research is
focusing to target other possible mechanisms that involve progress iron causing neu-
rodegeneration such as iron overloading neuroinflammation and oxidative stress. For
example, clinical trials of antioxidants usage such as Vitamin E and ω3 fatty acids
did not show any significant improvements in patients with neurodegenerative dis-
eases. Before developing a novel compound to treat iron causing neurodegeneration,
it must be needed profound investigations of underlying mechanisms of pathogenesis
of those neurological diseases. For example, disease-modifying approach has been
failed in stage III of the clinical trial for proving its efficacy against AD pathogene-
sis. It needs a better understanding of tau, Aβ, and other direct and indirect factors
to develop successful disease-modifying drugs. Altogether, new strategies need to
be focused to examine the neuroprotective activity of disease-modifying agents in
the presymptomatic stages of neurological diseases. In the development of drugs for
the treatment of these diseases, it is proposed to consider the joint use of drugs and
methods for regulating iron metabolism to improve the prevention and treatment of
diseases.
References
1. Andersen HH, Johnsen KB, Moos T (2014) Iron deposits in the chronically inflamed central
nervous system and contributes to neurodegeneration. Cell Mol Life Sci 71:1607–1622
2. Appleton TC, Morgan E, Baker E (1971) A morphological study of transferrin uptake by retic-
ulocytes. In: Trávníèek T, Neuwirt J (eds) The regulation of erythropoiesis and haemoglobin
synthesis. Universita karlova, prague, pp 310
3. Birkmayer W, Hornykiewicz O (1961) The L-3,4-dioxyphenylalanine (DOPA)-effect in
Parkinson-akinesia. Wien Klin Wochenschr 73:787–788
4. Busacchi V (1958) Vincenzo Menghini and the discovery of iron in the blood. Bull Sci Med
(bologna) 130:202–205
5. Charcot JM (1872) De la paralysie agitante. In: Oeuvres Complètes (t1) Leçons sur les maladies
du système nerveux, pp 155–188. Delahaye A, Paris: In: English: Charcot JM. 1877. On
Parkinson’s disease. In Lectures on diseases of the nervous system delivered at the Salpêtrière
(trans: Sigerson G). New Sydenham Society, London, pp 129–156
6. Chen H, Attieh ZK, Syed BA, Kuo YM, Stevens V, Fuqua BK, Andersen HS, Naylor CE,
Evans RW, Gambling L et al (2010) Identification of zyklopen, a new member of the verte-
brate multicopper ferroxidase family, and characterization in rodents and human cells. J Nutr
140:1728–1735
7. Curtis AR, Fey C, Morris CM, Bindoff LA, Ince PG, Chinnery PF, Coulthard A, Jackson MJ,
Jackson AP, McHale DP et al (2001) Mutation in the gene encoding ferritin light polypeptide
causes dominant adult-onset basal ganglia disease. Nat Genet 28:350–354
1 Brain Iron Metabolism and CNS Diseases 17
8. Conrad ME Jr, Crosby WH (1963) Intestinal mucosal mechanisms controlling iron absorption.
Blood 22:406–415
9. Conrad S, Genth H, Hofmann F, Just I, Skutella T (2007) Neogenin-RGMa signaling at the
growth cone is bone morphogenetic protein-independent and involves RHoA, ROCKk, and
PKC. J Biol Chem 282:16423–16433
10. Deane R, Zheng W, Zlokovic BV (2004) Brain capillary endothelium and choroid plexus
epithelium regulate transport of transferrin-bound and free iron into the rat brain. J Neurochem
88:813–820
11. Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt SJ, Moynihan J, Paw BH, Drejer A, Barut B,
Zapata A, Law TC, Brugnara C, Lux SE, Pinkus GS, Pinkus JL, Kingsley PD, Palis J, Fleming
MD, Andrews NC, Zon LI (2000) Positional cloning of zebrafish ferroportin1 identifies a
conserved vertebrate iron exporter. Nature 403:776–781
12. Drysdale JW, Munro HN (1965) Failure of actinomycin D to prevent induction of liver apofer-
ritin after iron administration. Biochim Biophys Acta 103:185–188
13. Ehringer H, Hornykiewicz O (1960) Verteilung von noradrenalin and dopamin im gehirn
des menschen und ihr verhalten bei erkrankungen des extrapyramidalen systems. Klin Wschr
38:1126–1239
14. Fenton HJH (1876) On a new reaction or tartaric acid. Chemical News 33:190
15. Fenton HJH (1893) The oxidation of tartaric acid in presence of iron. Chem Soc Proc 9:113
16. Fineberg RA, Greenberg DM (1955) Ferritin biosynthesis II. Acceleration of synthesis by the
administration of iron. J Biol Chem 214:97–106
17. Finch CA (1959) Body iron exchange in man. J Clin Invest 38:392–396
18. Fontés G, Thivolle I (1925) Sur la teneur du sérum en fer non hémoglobinique et sur sa
diminution au cours de l’anémie expérimentale. Compt Rend Soc Biol 96:687
19. Gowers WR (1899) Paralysis agitans. In: Allbutt A, Rolleston T (eds) A system of medicine.
Macmillan, London, pp 156–178
20. Guo B, Phillips JD, Yu Y, Leibold EA (1995) Iron regulates the intracellular degradation of
iron regulatory protein 2 by the proteasome. J Biol Chem 270:21645–21651
21. Hahn PF, Bale WF, Ross JF, Balfour WM, Whipple GH (1943) Radioactive iron absorption
by gastrointestinal tract: influence of anemia, anoxia, and antecedent feeding distribution in
growing dogs. J Exp Med 78:169–188
22. Hahn P, Qian Y, Dentchev T, Chen L, Beard J, Harris ZL et al (2004) Disruption of ceruloplasmin
and hephaestin in mice causes retinal iron overload and retinal degeneration with features of
age-related macular degeneration. Proc Natl Acad Sci U S A 101:13850–13855
23. Halliwell B, Gutteridge JM (1984) Role of iron in oxygen radical reactions. Methods Enzymol
105:47–56
24. Hentz MW, Kuhn LC (1996) Molecular control of vertebrate iron metabolism: mRNA-based
regulatory circuits operated by iron, nitric oxide, and oxidative stress. Proc Natl Acad Sci U S
A 93:8175–8182
25. Hentze MW, Muckenthaler MU, Galy B, Camaschella C (2010) Two to tango: regulation of
mammalian iron metabolism. Cell 142:24–38
26. Holmberg CG, Laurell CB (1945) Studies on the capacity of serum to bind iron—a contribution
to our knowledge of the regulation mechanism of serum iron. Acta Physiol Scand 10:307
27. Howard R, McShane R, Lindesay J, Ritchie C, Baldwin A, Barber R, Burns A, Dening T,
Findlay D, Holmes C, Hughes A, Jacoby R, Jones R, Jones R, McKeith I, Macharouthu A,
O’Brien J, Passmore P, Sheehan B, Juszczak E, Katona C, Hills R, Knapp M, Ballard C, Brown
R, Banerjee S, Onions C, Griffin M, Adams J, Gray R, Johnson T, Bentham P, Phillips P
(2012) Donepezil and meantime for moderate-to-severe Alzheimer’s disease. N Engl J Med
366:893–903
28. Hornykiewicz O (2002) Dopamine miracle: from brain homogenate to dopamine replacement.
Mov Disord 17:501–508
29. Holtz P, Heise R, Lüdtke K (1938) Fermentative degradation of l-dioxyphenylalanine (dopa)
by kidney. Naunyn-Schmiedeberg’s Arch. Pharmacol 191:87–118
30. Jandl JH, Katz JH (1963) The plasma-to-cell cycle of transferrin. J Clin Invest 42:314–326
18 A. Thirupathi and Y.-Z. Chang
31. Kasarskis EJ, Tandon L, Lovell MA, Ehmann WD (1995) Aluminum, calcium, and iron in the
spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe mass
spectroscopy: a preliminary study. J Neurol Sci 130:203–208
32. Ke Y, Qian ZM (2007) Brain iron metabolism: neurobiology and neurochemistry. Prog Neu-
robiol 83:149–173
33. Leibold EA, Munro HN (1988) Cytoplasmic protein binds in vitro to a highly conserved
sequence in the 5 untranslated region of ferritin heavy- and light-subunit mRNAs. Proc Natl
Acad Sci U S A. 85:2171–2175
34. Lecanu LR, l’hématosine de (1830) O matière colorante du sang. Ann Chim 45:5–27
35. Lecanu LR (1831) Nouvelle recherches sur le sang. Ann Chim 48:308–327
36. Li X, Jankovic J, Le W (2011) Iron chelation and neuroprotection in NDs. J Neural Trans
118:473–477
37. Manyam BV (1990) Paralysis agitans and levodopa in “Ayurveda”: ancient Indian medical
treatise. Mov Disord 5(1):47–48
38. Malecki EA, Devenyi AG, Beard JL, Connor JR (1999) Existing and emerging mechanisms
for transport of iron and manganese to the brain. J Neurosci Res 56:113–122
39. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Barrow D, Miret S, Bomford A, Peters
TJ, Farzaneh F, Hediger MA, Hentze MW, Simpson RJ (2000) A novel duodenal iron-regulated
transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Mol Cell
5:299–309
40. McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, Sager G, Mudaly E, Mudaly M, Richard-
son C, BarlowD Bomford A (2001) An iron-regulated ferric reductase associated with the
absorption of dietary iron. Science 291:1755–1759
41. Mayajima H (2003) Aceruloplasminemia an iron metabolic disorder. Neuropathology
23:345–350
42. Monarde N (1925) Joyful newes out of the newe founde worlde. New York
43. Morgan EH, Appleton TC (1969) Autoradiographic localization of 125-I-labelled transferrin
in rabbit reticulocytes. Nature 223:1371–1372
44. Mccance RA, Widdowson EM (1938) The absorption and excretion of iron following oral and
intravenous administration. J Physiol 94:148–154
45. Nicolas G, Bennoun M, Devaux I et al (2001) Lack of hepcidin gene expression and severe
tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad
Sci U S A. 98(15):8780–8785
46. Oshiro S, Kawahara M, Kuroda Y, Zhang C, Cai Y, Kitajima S, Shirao M (2000) Glial cells
contribute more to iron and aluminum accumulation but are more resistant to oxidative stress
than neuronal cells. Biochim Biophys Acta 1502:405–414
47. Owen D, Kühn LC (1987) Noncoding 3 sequences of the transfer in receptor gene are required
for mRNA regulation by iron. EMBO J 6:1287–1293
48. Parkinson J (1817) An essay on the shaking palsy. Whittingham and Rowland for Sherwood,
Needly and Jones, London
49. Pollycove M, Mortimer R (1961) The quantitative determination of iron kinetics and
hemoglobin synthesis in human subjects. J Clin Invest 40:753–782
50. Qian ZM, Chang YZ, Zhu L, Yang L, Du JR, Ho KP et al (2007) Development and
iron-dependent expression of hephaestin in different brain regions of rats. J Cell Biochem
102:1225–1233
51. Rouault TA (2006) The role of iron regulatory proteins in mammalian iron homeostasis and
disease. Nat Chem Biol 2:406–414
52. Schade AL, Caroline I (1944) Raw hen egg white and the role of iron in growth inhibition of
Shigella dysenteriae, Staphylococcus aureus, Escherichia coli and Saccharomyces cerevisiae.
Science 100:14–15
53. Santa-Maria I, Hernández F, Del Rio J, Moreno FJ, Avila J (2007) Tramiprosate, a drug of
potential interest for the treatment of Alzheimer’s disease, promotes an abnormal aggregation
of tau. Mol Neurodegener 6:17
1 Brain Iron Metabolism and CNS Diseases 19
54. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I (2004) Memantine
treatment in patients with moderate to severe Alzheimer disease already receiving donepezil:
a randomized controlled trial Memantine Study Group. JAMA 291(3):317–324
55. Troadec MB, Warner D, Wallace J et al (2011) Targeted deletion of the mouse Mitoferrin1
gene: from anemia to protoporphyria. Blood 117(20):5494–5502
56. Vanotti A, Delachaux A (1949) iron metabolism and its clinical significance. Grune & Stratton,
New York
57. Vivot RM, Goitia B, Usach V, Setton-Avruj PC (2013) DMT1 as a candidate for non-transferrin-
bound iron uptake in the peripheral nervous system. BioFactors 39:476–484
58. Vulpe CD, Kuo YM, Murphy TL, CowleyL Askwith C, Libina N, Gitschier J, Anderson
GJ (1999) Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is
defective in the sla mouse. Nat Genet 21:195–199
59. Wächtershäuser G (1990) Evolution of the first metabolic cycles. Proc Natl Acad Sci U S A
87:200–204
60. Wächtershäuser G (2000) Origin of life. Life as we don’t know it. Science 289:1307–1308
61. Wang SM, Fu LJ, Duan XL, Crooks DR, Yu P, Qian ZM, Di XJ, Li J, Rouault TA, Chang YZ
(2010) Role of hepcidin in murine brain iron metabolism. Cell Mol Life Sci 67(1):123–133
62. You LH, Yan CZ, Zheng BJ, Ci YZ, Chang SY, Yu P, Gao GF, Li HY, Dong TY, Chang YZ
(2017) Astrocyte hepcidin is a key factor in LPS-induced neuronal apoptosis. Cell Death Dis
8(3):e2676
63. Zahringer J, Baliga BS, Munro HN (1976) Novel mechanism for translational control in regu-
lation of ferritin synthesis by iron. Proc Natl Acad Sci USA 73:857–861
64. Zorzi G, Zibordi F, Chiapparini L, Bertini E, Russo L, Piga A et al (2011) Iron-related MRI
images in patients with pantothenate kinase-associated neurodegeneration (PKAN) treated with
deferiprone: results of a phase II pilot trial. Mov Disord 26:1756–1759
Chapter 2
Cellular Iron Metabolism and Regulation
Abstract Iron is an essential trace element in the human body, but excess iron is toxic
as it contributes to oxidative damage. To keep iron concentration within the optimal
physiologic range, iron metabolism at the cellular level and the whole systemic
level are tightly regulated. Balance of iron homeostasis depends on the expression
levels and activities of iron carriers, iron transporters, and iron regulatory and storage
proteins. Divalent metal transporter 1 (DMT1) at the apical membrane of intestinal
enterocyte brings in non-heme iron from the diet, whereas ferroportin 1 (FPN1) at the
basal membrane exports iron into the circulation. Plasma transferrin (Tf) then carries
iron to various tissues and cells. After binding to transferrin receptor 1 (TfR1), the
complex is endocytosed into the cell, where iron enters the cytoplasm via DMT1
on the endosomal membrane. Free iron is either utilized in metabolic processes,
such as synthesis of hemoglobin and Fe–S cluster, or sequestered in the cytosolic
ferritin, serving as a cellular iron store. Excess iron can be exported from the cell via
FPN1. The liver-derived peptide hepcidin plays a major regulatory role in controlling
FPN1 level in the enterocyte, and thus controls the whole-body iron absorption.
Inside the cells, iron regulatory proteins (IRPs) modulate the expressions of DMT1,
TfR1, ferritin, and FPN1 via binding to the iron-responsive element (IRE) in their
mRNAs. Both the release of hepcidin and the IRP–IRE interaction are coordinated
with the fluctuation of the cellular iron level. Therefore, an adequate and steady iron
supplement is warranted for the utilization of cells around the body. Investigations on
the molecular mechanisms of cellular iron metabolism and regulation could advance
the fields of iron physiology and pathophysiology.
Iron (Fe) is one of the essential trace metals in the human body, which is found in
the active centers of many enzymes and oxygen carrier proteins [1]. Iron is a critical
component of cytochromes a, b, and c, cytochrome oxidase, and iron–sulfur (Fe–S)
complexes of the oxidative chain, therefore important for adenosine triphosphate
(ATP) production. Iron is a co-enzyme in ribonucleoside reductase which commits to
DNA synthesis. Iron is also involved in TCA cycle since the succinate dehydrogenase
and aconitase are both Fe-dependent enzymes. In addition, the iron in the brain is
required for myelogenesis and myelin maintenance as well. Therefore, a constant
and readily available supply of iron is important for all the organs of the body [2].
The iron transport, storage, and regulation are tightly regulated at the cellular and
systemic levels [1, 2].
Human bodies contain 4–5 g of iron on average. Dietary iron is absorbed pre-
dominately in the duodenum and enters blood circulation in the small intestine. The
absorption of heme iron is poorly understood. Non-heme iron is transported across
the apical membrane of the intestinal enterocyte by divalent metal transporter 1
(DMT1) and is exported into the circulation via ferroportin 1 (FPN1). Once in blood
circulation, iron binds to apotransferrin and forms Fe-transferrin (Tf) complex [3].
Serum Tf is the major vehicle for iron transport in the body and carries iron to other
cells and tissues through the circulation [3]. At the target cell, Tf binds to transfer-
rin receptor 1 (TfR1) on the cell membrane, and the TfR1-Tf-Fe complex is then
endocytosed into the cell, where the iron is released [4]. Free iron either enters mito-
chondrion for utilization in metabolic processes, such as synthesis of hemoglobin
and Fe–S cluster, or is incorporated into the cytosolic iron storage protein, ferritin
and serves as a cellular store of iron [5]. Excess iron is transported out of the cell by
iron efflux protein ferroportin 1 (FPN1) located on the cell membrane [6].
About 2.5 g iron in the body is contained in the hemoglobin, the red pigment in red
blood cells, which is needed to carry oxygen through the blood. This portion of iron
is circulated in the body by macrophages after they engulf the senile erythrocytes and
release iron. A relatively small amount iron (3–4 mg) circulates through the plasma,
bound to Tf. Most of the rest iron is stored in ferritin complexes that are present in all
cells, but most common in bone marrow, liver, and spleen [1, 5]. Of these iron stores,
the iron in the liver is the primary physiologic source of reserve iron in the body.
There are a lower non-hemoglobin hemoproteins, such as myoglobin, cytochromes,
catalases, heme peroxidase, and endothelial nitric oxide synthase, involved in diverse
biological functions including the transportation of diatomic gases, chemical catal-
ysis, diatomic gas detection, and electron transfer [7]. Because of its toxicity, free
soluble iron in the body is kept in low concentration in the body.
2 Cellular Iron Metabolism and Regulation 23
Iron homeostasis in the body is regulated at two different levels, the systemic level
and the cellular level [8]. The systemic iron level is mainly balanced by the controlled
absorption of dietary iron by intestinal enterocytes, the cells that line at the interior of
the intestines. The cellular iron level is controlled differently by different cell types
due to the expression of particular iron regulatory and transport proteins [9].
At the systemic level, iron homeostasis involves the regulation of a peptide ‘hormone’
hepcidin [10]. Hepcidin is mainly produced by hepatocytes in response to high serum
iron concentration, inflammatory stimuli, or hypoxia [10, 11]. It binds to the extra-
cellular loop of FPN1 and causes its internalization and degradation, and thereby
reduces cellular iron efflux [2, 12]. Hepcidin regulates iron homeostasis mainly via
acting on FPN1 of intestinal enterocytes and FPN1 of macrophages, thereby affect-
ing iron intake from the intestine and iron release from macrophages [13, 14]. The
level of hepcidin is also regulated by the systemic iron level [15]. When body iron is
overloaded, the level of hepcidin increases to reduce circulating iron. When body iron
is deficient, the level of hepcidin decreases to promote small intestinal iron intake.
In addition to the iron level, inflammation, hypoxia, and erythropoiesis regulate the
expression of hepcidin as well [16].
that the IRPs knockout mice had significant misregulation of iron metabolism and
developed neurodegeneration [18, 19].
In the intestine, the control of ferritin and FPN1 expression by IRPs limits intesti-
nal iron absorption and thus controls the whole-body iron level [20]. In the bone
marrow, IRPs regulate the translation of erythroid 5-aminolevulinate synthase’s
mRNA; therefore, the erythroid heme biosynthesis is coordinated with iron avail-
ability. Besides, the translational control of HIF2α mRNA in the kidney by IRP1
coordinates erythropoietin synthesis with iron and oxygen supply [19, 20].
mRNA, bound to iron, which leads to the degradation of TfR1 mRNA by nuclease,
and subsequently decreases TfR1 protein synthesis [23].
Divalent metal transporter 1 (DMT1) is a bivalent metal ion transporter with a
molecular weight of approximately 61 kDa [24]. In 1997, both Harvard University
and Yale University reported this transmembrane iron transporter [24, 25]. DMT1
plays a key role in maintaining the homeostasis of bivalent metal ions in cells, espe-
cially iron and manganese [26]. DMT1 can be divided into DMT1 (+IRE) and DMT1
(−IRE) according to whether its 3 -UTR contains IRE sequence. DMT1 (+IRE) is
regulated by IRPs in response to intracellular iron level [18]. In the case of iron
overload, IRPs bind to iron, which reduces the stability of DMT1 (+IRE) mRNA,
promotes its degradation and decreases the level of DMT1 protein. On the contrary,
under iron deficiency status, DMT1 expression increases [18].
Ferroportin (FPN1), also known as MPT1 or IREG1, is the currently only known
multitransmembrane iron export protein in mammalian cells, with a molecular weight
of about 62 kDa [6]. Three groups independently discovered this iron transporter in
2000 [35–37]. FPN1 commonly expresses on the membrane of all the cells, which
allows Fe2+ to pass through. The basal surface of epithelial cells, the cellular surface of
macrophages in liver and spleen, and trophoblast of placenta express relatively higher
levels of FPN1, in order to export iron out of intestinal enterocytes, hepatocytes, and
splenic macrophages to the blood. FPN1 transports ferrous ions out of cells with
the assistance of ferrous oxidase ceruloplasmin (CP) or hephaestin (HP) [38]. The
expression of FPN1 is mainly regulated by two aspects. On the one hand, IRPs
regulate FPN1 expression at the post-transcriptional level via the IRP–IRE system.
The 5 -UTR of FPN1 mRNA has the IRE structure [39]. IRP–IRE binding weakens
the expression of FPN1 protein through translation inhibition. In addition, FPN1
protein level is also regulated by hepcidin. Hepcidin can recognize and bind to FPN1
on the membrane, which induces its internalization [14, 40]. After internalization,
FPN1 and hepcidin are transported to lysosomes and both were degraded [41]. It was
recently reported that hepcidin can also inhibit the activity of FPN1 [42, 43], which
then reduces the release of intracellular iron, thus causing changes in intracellular or
tissue iron levels. In addition, the regulation of hepcidin on FPN1 may also coordinate
with IRPs, together affecting FPN1’s level [44].
Iron regulatory proteins (IRPs) are cytosolic soluble RNA-binding proteins that bind
to IRE sequences on mRNAs of iron transporters and iron storage proteins, reg-
ulating their expression [17, 45]. IRPs have two forms, iron regulatory protein 1
(IRP1, ~90 kDa) and iron regulatory protein 2 (IRP2, ~105 kDa). IRP1 is rich in
most tissues, while IRP2 is most abundant in the brain and small intestine. When
iron is deficient in cells, IRPs bind to IRE of the target mRNA, and while iron
level is elevated, IRPs dissociate from IRE, regulating protein expression at the
post-transcriptional level [17, 23]. IRPs bound to IREs of TfR1 mRNAs, as well as
DMT1 mRNAs, promote their expression level, thus raising cellular iron absorp-
tion. Meanwhile, IRPs bound to IREs of FPN1 mRNAs reduce cellular iron release
by suppressing FPN1’s translation [39, 45]. Besides, IRPs regulate H-ferritin and
L-ferritin translation, thus adjusting iron storage. These mechanisms have probably
evolved to maintain the cytoplasmic LIP level at a steady level.
Hepcidin, a 25 amino acids peptide, produced mainly by the liver in response
to high serum iron, is the ‘master’ regulator of iron homeostasis at systemic level
[14]. Hepcidin was identified as a urinary antimicrobial peptide synthesized in the
2 Cellular Iron Metabolism and Regulation 27
liver in 2000 [10]. Hepcidin restricts the release of iron by controlling FPN1 on
the membrane, inducing its internalization and degradation [12]. Recently, the study
reported that hepcidin can also downregulate the activity of FPN1 [42]. Hepcidin
level is in turn regulated by body iron demand, through BMP/SMAD and TfR2
pathways. When the body iron is replete, a higher hepcidin concentration reduces
iron absorption at intestine and impairs iron release from iron stores; but when the
body iron is deficient, hepcidin level is low, thereby favoring iron absorption and
delivery to the plasma from storage sites [15]. Inflammation is also an important factor
affecting the expression of hepcidin. Interleukin-6 (IL-6) upregulates the expression
of hepcidin through the STAT3 pathway [46]. In addition, hepcidin is negatively
regulated by erythropoietin, oxygen environment, and sex hormones [16, 47].
Iron deficiency is a state that the body iron amount is lower than the normal physio-
logic conditions, and the body lacks enough iron to supply its needs. Iron deficiency
may impair the synthesis of essential iron-containing proteins that are required for
normal cellular physiology and result in a range of adverse consequences [48]. When
the loss of iron is not sufficiently compensated by adequate iron intake from the diet,
a state of iron deficiency will develop over time [48].
Untreated iron deficiency can lead to iron deficiency anemia, the most common
type of anemia [49]. In iron deficiency anemia, the body lacks sufficient amounts of
iron to produce the protein hemoglobin, thereby resulting in inadequate red blood
cells (RBCs). Besides, iron deficiency can also be resulted from genetic disturbances
in iron homeostasis. In particular, mutations in the TMPRSS6 gene, which encodes
an upstream regulator of hepcidin, can lead to iron-refractory iron deficiency anemia
[50]. The treatment of mild iron deficiency can be achieved simply by increasing the
amount of bioavailable iron in the diet or by taking iron supplements; while in the
severe case of iron deficiency anemia, an intravenous iron infusion may be used to
efficiently deliver a large amount of iron [51].
The anemia of chronic disease is the second most common form of anemia that is
caused by iron deficiency with inflammation [52]. The anemia of chronic disease is
usually due to the presence of chronic infection, chronic immune activation, malig-
nancy, or various chronic inflammatory states. These conditions all produce massive
elevation of pro-inflammatory cytokine IL-6. IL-6 stimulates hepcidin production
and release from the liver, which in turn reduces the iron efflux protein FPN1, result-
ing in reduced iron uptake at intestine and reduced iron release from iron stores and
macrophages, thereby reducing iron in the circulation. The plasma iron concentra-
tion decreases, and macrophages and other cell types sequester iron. If this condition
persists, the iron supply to the erythroid marrow can be compromised, resulting in
28 G. Gao et al.
reduced erythropoiesis, namely the anemia of chronic disease [52]. Hepcidin antago-
nists or agents that block hepcidin gene synthesis and subsequently increase intestinal
iron absorption have shown promise in the treatment of the anemia of inflammation
[53].
Iron overload is also associated with a variety of diseases [54]. Excess LIP in the
cell may catalyze reactions that generate ROS and consequently cause oxidative
damage to cells and tissues, which can finally lead to tissue fibrosis and organ dys-
functions [55]. Some iron overload diseases are caused by genetic mutations in
the genome, such as hemochromatosis, atransferrinemia, aceruloplasminemia, and
Friedreich ataxia [54–56]. Some are resulted from the chronic disorders, e.g., chronic
liver disease [57]. Iron overload is also complicated with a range of other diseases,
such as cystic fibrosis, fatty liver disease, and neurodegenerative diseases, including
Alzheimer’s disease (AD), Parkinson’s disease (PD), and so on [54, 58].
Hemochromatosis is a major group of primary iron overload diseases, which is
resulted from mutations in genes that are involved in the transport of iron or the regula-
tion of iron metabolism [59]. Currently, there are five main types of hemochromatosis
are identified due to mutations in the genes encoding hemochromatosis protein HFE,
hemojuvelin (HJV), hepcidin, TfR2, and FPN1 [59, 60]. The common feature of these
different types is a reduced hepcidin expression level [59]. The reduced hepcidin
level leads to a higher FPN1 level on the membrane of intestinal enterocytes, which
increases the absorption of dietary iron, and therefore the body iron load increases.
In these patients, iron accumulation was observed in many organs, particularly in the
liver, heart, and pancreas [54]. Clinical syndromes of the hemochromatosis patients
include hepatic fibrosis and cirrhosis, hepatocellular carcinoma, arthritis, cardiomy-
opathy, and so on [59]. The disease of hemochromatosis is relatively common in the
populations of Northern European origin, but is a rare case in other populations [60].
Hemochromatosis is typically treated via phlebotomy with the removal of ∼0.5 g
Fe/L blood, which is the currently most effective and relatively inexpensive method
[54]. After removal, the serum iron and RBCs in the blood, iron in the storage sites,
notably in the liver, will be subsequently released to replenish the reduced RBCs in
blood for erythropoiesis [54].
Thalassemia is one kind of anemia with iron overload instead of iron deficiency.
In thalassemia, patients have defects in either the α- or β-globin chain, causing the
production of abnormal hemoglobin, thereby characterized by reduced RBCs and
thus anemia [61]. However, blood transfusions for treatment of this inherited disor-
der usually lead to secondary iron overload, with resulting heart or liver diseases,
infections, and osteoporosis. It is most common among people of Italian, Greek, Mid-
dle Eastern, South Asian, and African descent. In the treatment of iron overload in
thalassemias, iron chelators are commonly used [61]. Hepcidin mimetics or agents
2 Cellular Iron Metabolism and Regulation 29
that increase hepcidin expression could also be promising drugs in the treatment of
hemochromatosis and thalassemia [62].
Abnormal iron accumulation has been detected in various neurological diseases in
CNS, including neurodegeneration with brain iron accumulation (NBIA) diseases,
AD, PD, Friedreich’s ataxia, amyotrophic lateral sclerosis, stroke, and so on [63,
64]. Although the causes of most of the neurological disorders have not been fully
understood, common features of these diseases are iron accumulation and increased
oxidative damage [63]. Iron chelators, such as deferoxamine (DFO), have been shown
good efficacy as potential therapeutic agents for iron overload-associated neurolog-
ical disorders [65]. As early as in 1991, McLachlan et al. had used DFO to treat
AD in a clinical trial. They found that continuous administration of DFO could alle-
viate AD-induced cognitive impairment [66]. In the recent years, it was found that
DFO treatment reduced the cognitive decline and the accumulation of amyloid-β in
APP/PS1 mice, which may be related to the induction of M2 activation and inhibition
of M1 activation by DFO in the hippocampus of mice [67]. Researchers have suc-
cessfully developed a brain-targeting peptide-modified nanopolymer encapsulated
DFO, which can cross the blood–brain barrier through receptor-mediated endocy-
tosis [68]. This improvement greatly increased the efficiency of DFO entering the
brain, and significantly decreased the iron content in substantia nigra and striatum,
reduced oxidative stress level, and alleviated dopaminergic neuron damage in PD
mouse model [68].
2.5 Conclusions
Iron is an essential trace element in human body, but it is also toxic in excess. Thus,
iron homeostasis at cellular level and the whole-body iron concentration are tightly
regulated to keep it within the optimal physiologic range. Body iron is acquired from
the diet and exported into the circulation for utilization. Therefore, the amount of iron
uptake at intestinal enterocyte is fairly critical. Both the levels of DMT1 and FPN1
expressed by intestinal enterocytes are must under precise regulations. The liver-
derived peptide hepcidin in serum plays a major regulatory in controlling FPN1
level on the membrane of intestinal enterocytes. While intracellularly, IRPs regulate
the levels of DMT1, TfR1, ferritin and FPN1 in response to iron level changes.
Therefore, an adequate and steady iron supplement is warranted for the utilization of
cells around the body. Similar as the iron regulation in enterocyte, the cellular iron
levels in other cells and tissues are controlled differently by different cell types due to
the expression of particular iron regulatory and transport proteins. Abnormal levels
of body iron, too little or too much, are commonly associated with diseases of iron
deficiency or iron overload. Despite major recent advances have achieved regarding
the mechanisms of cellular iron metabolism, much remains to be learned about iron
physiology and pathophysiology.
30 G. Gao et al.
References
1. Muckenthaler MU, Rivella S, Hentze MW, Galy B (2017) A red carpet for iron metabolism.
Cell 168(3):344–361
2. Anderson GJ, Wang F (2012) Essential but toxic: controlling the flux of iron in the body. Clin
Exp Pharmacol Physiol 39(8):719–724
3. Aisen P, Leibman A, Zweier J (1978) Stoichiometric and site characteristics of the binding of
iron to human transferrin. J Biol Chem 253(6):1930–1937
4. Huebers HA, Finch CA (1987) The physiology of transferrin and transferrin receptors. Physiol
Rev 67(2):520–582
5. Harrison PM, Arosio P (1996) The ferritins: molecular properties, iron storage function and
cellular regulation. Biochim Biophys Acta 1275(3):161–203
6. Drakesmith H, Nemeth E, Ganz T (2015) Ironing out ferroportin. Cell Metab 22(5):777–787
7. Paoli M, Marles-Wright J, Smith A (2002) Structure-function relationships in heme-proteins.
DNA Cell Biol 21(4):271–280
8. Hentze MW, Muckenthaler MU, Galy B, Camaschella C (2010) Two to tango: regulation of
Mammalian iron metabolism. Cell 142(1):24–38
9. Aisen P (1992) Entry of iron into cells: a new role for the transferrin receptor in modulating
iron release from transferrin. Ann Neurol 32(Suppl):S62–S68
10. Park CH, Valore EV, Waring AJ, Ganz T (2001) Hepcidin, a urinary antimicrobial peptide
synthesized in the liver. J Biol Chem 276(11):7806–7810
11. Nicolas G, Chauvet C, Viatte L, Danan JL, Bigard X, Devaux I et al (2002) The gene encoding
the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin
Invest 110(7):1037–1044
12. Ramey G, Deschemin JC, Durel B, Canonne-Hergaux F, Nicolas G, Vaulont S (2010) Hepcidin
targets ferroportin for degradation in hepatocytes. Haematologica 95(3):501–504
13. Beutler E (2004) Cell biology. “Pumping” iron: the proteins. Science 306(5704):2051–2053
14. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM et al (2004) Hepcidin
regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science
306(5704):2090–2093
15. Parrow NL, Fleming RE (2014) Bone morphogenetic proteins as regulators of iron metabolism.
Annu Rev Nutr 34(34):77–94
16. Ganz T (2011) Hepcidin and iron regulation, 10 years later. Blood 117(17):4425–4433
17. Rouault TA (2006) The role of iron regulatory proteins in mammalian iron homeostasis and
disease. Nat Chem Biol 2(8):406–414
18. Leipuviene R, Theil EC (2007) The family of iron responsive RNA structures regulated by
changes in cellular iron and oxygen. Cell Mol Life Sci 64(22):2945–2955
19. Meyron-Holtz EG, Ghosh MC, Rouault TA (2004) Mammalian tissue oxygen levels modulate
iron-regulatory protein activities in vivo. Science 306(5704):2087–2090
20. Anderson GJ, Frazer DM (2017) Current understanding of iron homeostasis. Am J Clin Nutr
106(Suppl 6):1559S–1566S
21. Bali PK, Zak O, Aisen P (1991) A new role for the transferrin receptor in the release of iron
from transferrin. Biochemistry 30(2):324–328
22. Cheng Y, Zak O, Aisen P, Harrison SC, Walz T (2004) Structure of the human transferrin
receptor-transferrin complex. Cell 116(4):565–576
23. Wallander ML, Leibold EA, Eisenstein RS (2006) Molecular control of vertebrate iron home-
ostasis by iron regulatory proteins. Biochim Biophys Acta 1763(7):668–689
24. Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF, Boron WF et al (1997)
Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature
388(6641):482–488
25. Fleming MD, Trenor CC, Su MA, Foernzler D, Beier DR, Dietrich WF et al (1997) Micro-
cytic anaemia mice have a mutation in Nramp2, a candidate iron transporter gene. Nat Genet
16(4):383–386
2 Cellular Iron Metabolism and Regulation 31
26. Garrick MD, Dolan KG, Horbinski C, Ghio AJ, Higgins D, Porubcin M et al (2003) DMT1: a
mammalian transporter for multiple metals. Biometals 16(1):41–54
27. Theil EC (1987) Ferritin: structure, gene regulation, and cellular function in animals, plants,
and microorganisms. Annu Rev Biochem 56:289–315
28. Arosio P, Ingrassia R, Cavadini P (2009) Ferritins: a family of molecules for iron storage,
antioxidation and more. Biochim Biophys Acta 1790(7):589–599
29. Levi S, Corsi B, Bosisio M, Invernizzi R, Volz A, Sanford D et al (2001) A human mitochondrial
ferritin encoded by an intronless gene. J Biol Chem 276(27):24437–24440
30. Corsi B, Cozzi A, Arosio P, Drysdale J, Santambrogio P, Campanella A et al (2002) Human mito-
chondrial ferritin expressed in HeLa cells incorporates iron and affects cellular iron metabolism.
J Biol Chem 277(25):22430–22437
31. Nie G, Sheftel AD, Kim SF, Ponka P (2005) Overexpression of mitochondrial ferritin causes
cytosolic iron depletion and changes cellular iron homeostasis. Blood 105(5):2161–2167
32. Santambrogio P, Biasiotto G, Sanvito F, Olivieri S, Arosio P, Levi S (2007) Mitochondrial
ferritin expression in adult mouse tissues. J Histochem Cytochem 55(11):1129–1137
33. Cocco E, Porrini V, Derosas M, Nardi V, Biasiotto G, Maccarinelli F et al (2013) Protective
effect of mitochondrial ferritin on cytosolic iron dysregulation induced by doxorubicin in HeLa
cells. Mol Biol Rep 40(12):6757–6764
34. Wang L, Yang H, Zhao S, Sato H, Konishi Y, Beach TG et al (2011) Expression and localization
of mitochondrial ferritin mRNA in Alzheimer’s disease cerebral cortex. PLoS ONE 6(7):e22325
35. Abboud S, Haile DJ (2000) A novel mammalian iron-regulated protein involved in intracellular
iron metabolism. J Biol Chem 275(26):19906–19912
36. Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt SJ, Moynihan J et al (2000) Posi-
tional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter. Nature
403(6771):776–781
37. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Barrow D et al (2000) A novel duo-
denal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the
circulation. Mol Cell 5(2):299–309
38. Knutson MD, Oukka M, Koss LM, Aydemir F, Wessling-Resnick M (2005) Iron release from
macrophages after erythrophagocytosis is up-regulated by ferroportin 1 overexpression and
down-regulated by hepcidin. Proc Natl Acad Sci USA 102(5):1324–1328
39. Lymboussaki A, Pignatti E, Montosi G, Garuti C, Haile DJ, Pietrangelo A (2003) The role
of the iron responsive element in the control of ferroportin1/IREG1/MTP1 gene expression. J
Hepatol 39(5):710–715
40. Qiao B, Sugianto P, Fung E, del-Castillo-Rueda A, Moran-Jimenez MJ, Ganz T et al (2012)
Hepcidin-induced endocytosis of ferroportin is dependent on ferroportin ubiquitination. Cell
Metab 15(6):918–924
41. Preza GC, Pinon R, Ganz T, Nemeth E (2013) Cellular catabolism of the iron-regulatory peptide
hormone hepcidin. PLoS ONE 8(3):e58934
42. Zhang DL, Rouault TA (2018) How does hepcidin hinder ferroportin activity? Blood
131(8):840–842
43. Zhang DL, Wu J, Shah BN, Greutelaers KC, Ghosh MC, Ollivierre H et al (2018) Erythro-
cytic ferroportin reduces intracellular iron accumulation, hemolysis, and malaria risk. Science
359(6383):1520–1523
44. Wang L, Liu X, You LH, Ci YZ, Chang S, Yu P et al (2019) Hepcidin and iron regulatory
proteins coordinately regulate ferroportin 1 expression in the brain of mice. J Cell Physiol
234(5):7600–7607
45. Pantopoulos K (2004) Iron metabolism and the IRE/IRP regulatory system: an update. Ann N
Y Acad Sci 1012:1–13
46. Nemeth E, Valore EV, Territo M, Schiller G, Lichtenstein A, Ganz T (2003) Hepcidin, a putative
mediator of anemia of inflammation, is a type II acute-phase protein. Blood 101(7):2461–2463
47. Vokurka M, Krijt J, Sulc K, Necas E (2006) Hepcidin mRNA levels in mouse liver respond to
inhibition of erythropoiesis. Physiol Res 55(6):667–674
48. Zimmermann MB, Hurrell RF (2007) Nutritional iron deficiency. Lancet 370(9586):511–520
32 G. Gao et al.
49. Naigamwalla DZ, Webb JA, Giger U (2012) Iron deficiency anemia. Can Vet J-Revue Vet Can
53(3):250–256
50. Finberg KE, Heeney MM, Campagna DR, Aydinok Y, Pearson HA, Hartman KR et al (2008)
Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat Genet
40(5):569–571
51. Powers JM, Buchanan GR (2014) Diagnosis and management of iron deficiency anemia. Hema-
tol Oncol Clin North Am 28(4):729–745, vi–vii
52. Peng YY, Uprichard J (2017) Ferritin and iron studies in anaemia and chronic disease. Ann
Clin Biochem 54(1):43–48
53. Camaschella C (2017) New insights into iron deficiency and iron deficiency anemia. Blood
Rev 31(4):225–233
54. Fleming RE, Ponka P (2012) Iron overload in human disease. N Engl J Med 366(4):348–359
55. Anderson GJ (2007) Mechanisms of iron loading and toxicity. Am J Hematol 82(12):1128–1131
56. Doyle A, Rusli F, Bhathal P (2015) Aceruloplasminaemia: a rare but important cause of iron
overload. BMJ Case Rep 2015
57. Kew MC (2014) Hepatic iron overload and hepatocellular carcinoma. Liver Cancer 3(1):31–40
58. Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L (2014) The role of iron in brain ageing
and neurodegenerative disorders. Lancet Neurol 13(10):1045–1060
59. Pietrangelo A (2010) Hereditary hemochromatosis: pathogenesis, diagnosis, and treatment.
Gastroenterology 139(2):393–408, 408 e391–e392
60. Barton JC, Edwards CQ, Acton RT (2015) HFE gene: structure, function, mutations, and
associated iron abnormalities. Gene 574(2):179–192
61. Taher AT, Weatherall DJ, Cappellini MD (2018) Thalassaemia. Lancet 391(10116):155–167
62. Olivieri NF, Brittenham GM (2013) Management of the thalassemias. Cold Spring Harb Per-
spect Med 3(6)
63. Ndayisaba A, Kaindlstorfer C, Wenning GK (2019) Iron in neurodegeneration—cause or con-
sequence? Front Neurosci 13:180
64. Rouault TA (2013) Iron metabolism in the CNS: implications for neurodegenerative diseases.
Nat Rev Neurosci 14(8):551–564
65. Dusek P, Schneider SA, Aaseth J (2016) Iron chelation in the treatment of neurodegenerative
diseases. J Trace Elem Med Biol 38:81–92
66. Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W et al (1991) Intra-
muscular desferrioxamine in patients with Alzheimer’s disease. Lancet 337(8753):1304–1308
67. Zhang Y, He ML (2017) Deferoxamine enhances alternative activation of microglia and inhibits
amyloid beta deposits in APP/PS1 mice. Brain Res 1677:86–92
68. You L, Wang J, Liu T, Zhang Y, Han X, Wang T et al (2018) Targeted brain delivery of rabies
virus glycoprotein 29-modified deferoxamine-loaded nanoparticles reverses functional deficits
in parkinsonian mice. ACS Nano 12(5):4123–4139
Chapter 3
Brain Iron Metabolism and Regulation
Abstract With the development of research, more and more evidences suggested
that mutations in the genes associated with brain iron metabolism induced diseases in
the brain. Brain iron metabolism disorders might be one cause of neurodegenerative
diseases. This review mainly summarizes the normal process of iron entry into the
brain across the blood–brain barrier, and the distribution and transportation of iron
among neurons and glial cells, as well as the underlying regulation mechanisms. To
understand the mechanisms of iron metabolism in the brain will provide theoretical
basis to prevent and cure brain diseases related to iron metabolism disorders.
3.1 Introduction
Iron is one of the most abundant trace elements and plays great roles in keeping
physiological homeostasis in the body. In the central nervous system (CNS), iron
participates in the synthesis of myelin and neurotransmitters [24, 39]. Iron deficiency
in the brain of infants will lead to abnormal neural development and mental retarda-
tion [21, 70]. While iron accumulation in specific brain regions is also shown to be
related to the patients with neurodegenerative diseases such as Parkinson’s Disease
(PD), Alzheimer’s Disease (AD) and Huntington’s Disease (HD). Excess iron can
react with cellular H2 O2 and generate reactive oxygen species (ROS) through Fenton
reaction, and increased ROS could induce a cascade of events to destroy membrane,
protein, and nucleic acid, which caused apoptosis and even cell death [38, 65, 96].
Therefore, it is very important to keep iron homeostasis in the brain.
Iron homeostasis in the brain was regulated strictly in mammalian animals, and it
depends on BBB and many molecules involved in the transportation of iron from the
blood circulation. In CNS, the microvascular endothelial cell (BMVEC), together
with astrocyte end-feet and pericytes, forms BBB which is the primary gatekeeper
for iron entry into the brain. Because there are tight junctions between BMVEC,
iron was first uptake into BMVEC from blood and then across BBB with the help of
many iron metabolism-related proteins [1, 44].
Long time ago, it is thought that iron can cross BBB into the brain mainly during
infancy before the formation of BBB. Now, new data demonstrated that iron can also
enter the brain across BBB in the adult brain, which depends on transferrin receptor
1 (TfR1) [56] and ferroportin 1 (FPN1) [50, 71, 92]. Iron can be bound in transferrin
(Tf) and transport in the blood, and Tf-TfR1 has been known as the main major route
of iron transport across the luminal membrane of the capillary endothelium from the
blood into endothelial cells [8, 47, 56]. First, Tf-bound iron in serum reached BBB,
the Tf-Fe binds to TfR1 in BMVEC, which forms endosomes after endocytosis of Tf-
TfR1. When pH value decreased to 5.5~6.5 in the endosomes, iron dissociated from
Tf and reduced to ferrous iron which released from the endosomes into endothelial
cytoplasm with the help of divalent metal transporter 1 (DMT1) [80, 81]. After
dissociation of iron from Tf-TfR1, apo-Tf-TfR1 returned to the apical surface of
BMVEC for the next round of iron uptake [9, 74], and intracellular iron is then either
incorporated into functional proteins, stored in ferritin, or continued to transport
across the abluminal membrane of the endothelial cell into the brain interstitium and
parenchyma [8, 56]. During the process of iron transport across the basal membrane
of intestinal epithelial cells, ferroportin 1 (FPN1) is an iron exporter and responsible
for iron efflux from epithelia into the blood in the presence of hephaestin (HP) and
ceruloplasmin (CP). Our previous results showed that FPN1 was distributed in murine
brain such as cerebral cortex, hippocampus, and striatum [88]. Others also reported
that FPN1 was also expressed in BBB, ependymocytes and choroid plexus, and that
FPN1 was especially expressed in the basal surface of BMVEC [90]. Moreover, CP
was also mainly expressed in the end-feet of astrocytes surrounding microvascular
vessels in the brain [41, 42, 68]. These evidences suggested that FPN1/CP might
play critical roles in iron efflux into brain parenchyma from BMVEC just similar to
that of enterocytes in duodenum. It is also reported that astrocytes can also directly
3 Brain Iron Metabolism and Regulation 35
uptake ferrous iron from BMVEC through the end-feet [49, 64]. However, there are
no direct evidences in vivo.
Besides Tf-TfR1 dependent iron uptake pathway, there are Tf-independent iron
transport ways. Ferritin (H-ferritin) can also deliver iron to the brain across BBB,
which is an iron transport system independent of Tf-TfR system. Ferritin might bind
to ferritin receptor or be transcytosed across the BBB [24]. Lactoferrin (Lf) and
its receptor (LfR), glycosylphosphatidylinositol anchored form of melanotransferrin
(p97, MTf), and soluble form of MTf also play roles in iron uptake across BBB from
the blood [22, 59]. Lf belongs to the Tf family and it is an iron-binding glycoprotein
which is distributed in milk or other secretion in peripheral tissues. In CNS, Lf is
expressed and secreted from activated microglia [2, 23], and LfR is expressed in
neurons, BMVEC, and some glial cells. Lf-LfR might play similar roles as Tf-TfR1
in iron transport into the brain parenchyma [21]. MTf is a transferrin homolog, and
GPI-MTf also plays similar roles as Tf-TfR1 in iron transport across BBB, and
secreted MTf seems to play more important roles in iron metabolism of patients
[59].
However, it is still not fully understood the detailed regulatory mechanisms of the
crosstalk between brain iron uptake and release at the BBB in vivo.
It is speculated that iron cross the blood–cerebrospinal fluid barrier (BCB) from
the blood is similar to iron transport at BBB. In situ hybridization found that the
choroid plexus expressed high levels of TfR1, DMT1, FPN1, Dcytb, and soluble
form of CP and HP, which suggested that choroid plexus might mediate significant
iron transport in CNS [52, 67, 78]. The choroid plexus can synthesize and secret
Tf, which might be involved in iron transport from BCB to the brain. But choroid
plexus-derived Tf does not seem too important to bind iron and transport iron in the
brain interstitium because [59 Fe125 I]Tf injected intracerebroventricularly was never
observed in regions distant from the CSF. Tf in the CSF probably plays a significant
role in export to the blood of iron [55].
Stromal cell-derived receptor 2 (SDR2) is expressed in the choroid plexus and
ependymal cells lining the four ventricles with in situ hybridization analysis [85].
SDR2 is the homolog of duodenal cytochrome b that is a ferric reductase, and DMT1
is also located in the choroid plexus [29], so SDR2 might also have ferric reductase
activity and participate in iron uptake with DMT1 at BCB.
36 P. Yu and Y.-Z. Chang
After iron is released into brain interstitial fluid, it is distributed and transported
among different cell types in different brain regions. It is reported that 10% of non-
heme iron entered the brain across BBB from the vertebrate blood and that iron levels
are different in different brain regions [11, 77]. Iron contents are most abundant in
the extrapyramidal system, especially in the basal ganglia region [58, 60], and iron
concentrations are high in subtantia nigra (SN), red nucleus, and cerebellar dentate
gyrus, while iron is relatively low in cerebral cortex, and iron levels are the lowest
in white matter and medulla oblongata [17, 58, 60].
Iron is released into the brain interstitial fluid across BBB, and it is absorbed by
neurons and different types of glial cells, but iron is distributed unevenly in these
cells [57, 77]. Neuroglia expressed almost all the iron transporters and distinct profiles
of iron metabolism related proteins [6, 32, 78, 89]. Data from primary cultures of
neurons, astrocytes, and microglia showed that iron retention in microglia was three
times higher than that of neurons when they were incubated with iron [7]. Studies in
a hippocampal slice culture mode also supported that ferritin mRNA was induced by
iron predominantly in microglia and oligodendrocytes, followed by astrocytes, but
never in neurons [31]. So neuroglia are indispensable to maintain iron homeostasis
and normal physiological function in the brain.
As for neurons, they express TfR1 and acquire iron through classical Tf-TfR1-
dependent iron uptake pathway from the brain interstitial fluid [30]. Neurons can also
uptake ferrous and ferric iron through DMT1 and trivalent cation-specific transporter
(TCT1), respectively [4, 39, 73].
As for astrocytes, the major glial cell type in CNS, they are important components
of BBB, so they play crucial roles in iron transport and are responsible for iron distri-
bution in the brain [10, 57]. They exclusively express glycosylphosphatidylinositol-
anchored CP (GPI-CP) in the end-feet surrounding BBB [35, 67]. Astrocytes also
secrete a soluble form of CP into interstitium which also facilitates iron trafficking
across BBB into the brain [52, 83]. Both GPI-CP and soluble form of CP are required
for the stability of FPN1 in the membrane and iron release from BMVEC. Moreover,
hephaestin present in astrocytes [10, 86] can also oxidize BMVEC released ferrous
iron to ferric iron so as to facilitate incorporation of iron into Tf and the following
transportation [13, 79, 94]. Astrocytes acquire iron across the interstitial space using
3 Brain Iron Metabolism and Regulation 37
NTBI mechanisms such as citrate, ATP, ascorbic acid, DMT1, and Zip [45, 69, 91].
They release iron through FPN1, and the released iron can be acquired by neurons
and other types of glial cells.
Oligodendrocytes are unique to the brain, because they are the only cells with
synthesized Tf and releasing Tf, and they express the Tim2 (T cell immunoglobulin
and mucin domain protein-2) ferritin transporter and contain a large amount of the
brain’s lipids and cholesterol [70], and they have high levels of iron and require large
amounts of iron in the myelination of neuronal axons to form the white matter in the
brain [5].
Microglia compromise approximately 12% of cells in the brain, and the resident
microglia are very stable. Microglia express the transporters or molecules involved in
iron metabolism, and they are most efficient in accumulating iron as mentioned above
[7, 83]. Under inflammation and environmental and endogenous stimuli, microglia
can sense and recognize the changes, and then, they become activated, iron retention
and affect the iron metabolism and function of neurons [83, 84]. Immunofluorescence
and hybridization in situ results revealed that activated microglia could synthesize
and secrete lactoferrin [2, 23], which can also affect the iron metabolism of lactoferrin
receptor expressing cells through the receptor-mediated pathway [91].
After iron is used for neuronal and glial metabolism and stored in ferritin, excess
iron is released via FPN1. The excess iron can be exported back to the interstitial
fluid or released into the cerebrospinal fluid in the brain ventricles [8, 27, 97], and the
choroidal epithelia capture iron by TfR1-dependent or TfR1-independent pathway
and then transport it back to the blood circulation [27].
Iron homeostasis is very important for normal brain functions. Iron homeostasis
depends on iron uptake, storage, and release [34]. Iron regulatory proteins (IRPs)
and hepcidin are critical regulators and coordinate with each other to maintain iron
homeostasis [28, 87].
IRP1 share high sequence homology with IRP2, and IRPs will regulate iron uptake,
iron storage, and iron release proteins with iron responsive element (IRE) in the gene
transcripts [61]. When cellular iron levels change, IRPs will regulate iron metabolism
through IRE-IRP regulatory systems at post-transcription level [40, 66].
In 2006, Rouault lab reported that IRP1−/− IRP2−/− double knockout mice were
embryonically lethal [82], which demonstrated the critical roles of IRPs in life.
38 P. Yu and Y.-Z. Chang
However, IRP1 and IRP2 have different distribution patterns, only IRP2 is highly
expressed in the brain [54]. Rouault lab and others also generated IRP2−/− mice,
and it was reported that excessive iron is accumulated in the brain of IRP2−/− mice
and neurodegenerative diseases like impairments were observed in aging IRP2−/−
mice [12, 25, 46, 66, 98], which further confirmed the crucial role of IRP2 in the
regulation of brain iron metabolism.
When cellular iron concentrations change, the expression of IRP2 will be altered
accordingly. Decreased cellular iron contents can increase the expression of IRP2,
which can bind to IRE in 5 -untranslated region (UTR) of the mRNA such as ferritin
mRNA, IRE in 3 UTR of transferrin receptor 1 (TfR1) mRNA, and DMT1 with IRE
[DMT1 (+IRE)] mRNA, thus induce the inhibition of ferritin expression, elevation
of TfR1 and DMT1 (+IRE) expression, which increase iron uptake and decrease iron
storage to relieve the iron deficiency in the cell. The vice is versa [20, 54, 61, 66,
93]. Therefore, IRE-IRP system can maintain iron homeostasis responding to altered
cellular iron levels in the brain.
sion of TfR1, DMT1 and FPN1, and hepcidin inhibits TfR1 expression via a cyclic
AMP-protein kinase A pathway, which implies the existence of a novel hepcidin-
receptor on the membrane of astroglia [18, 83]. However, there are no direct evi-
dences to show that brain hepcidin regulates the content of brain iron through effects
on FPN1 of BMVEC in vivo.
In fact, hepcidin mRNA levels in the brain have been shown to be much lower
than those in non-neural tissues or blood by qPCR experiments [88], but most
researches draw the conclusions of hepcidin-mediated regulation of FPN1 expres-
sion in BMVECs or neurons based on the usage of a considerably high concentration
of supraphysiological hepcidin (approximately 20-fold higher than that in blood).
Based on these data and facts, it is still to be elucidated how the low concentration
of hepcidin in the brain can regulate FPN1 in BMVECs and other types of cells.
It is worth noting that hepcidin and IRE-IRP systems must have communications
and coordination in their regulation of brain iron metabolisms [87]. In addition,
erythroferrone (ERFE) is a newly identified protein by Ganz group, and ERFE could
inhibit hepcidin and thus regulate iron metabolism in response to erythropoietin
stimulation under the stress condition [36, 37]. ERFE is also detected in the brain
with real-time PCR [37], but it is still unclear about its regulation of brain iron
metabolism. It is also unknown if there is any other functions of ERFE in the brain,
which needs to be further investigated.
3.5 Conclusion
Although brain iron metabolism and its regulation are complicated and there are still
some puzzles to be unraveled, the regulation profile is becoming clearer for us. Iron
is transported in the blood and entered the brain across BBB. Then iron is acquired by
neurons and different types of glial cells through iron uptake proteins, and released
from these cells through FPN1. Brain iron homeostasis is coordinately regulated by
IRP2 and hepcidin at post-transcriptional and protein levels, respectively. However,
there are still some problems to be answered and understood, such as how the iron
was released from the brain? Is the recently concerned glymphatic pathway involved
in it [76]? Which kind of signal initiates the uptake of iron and release from the
brain? The answers to these problems will not only enrich the theories of brain
iron metabolism and regulation, but also provide strategies to prevent and treat iron
metabolism disorders and related neurodegenerative diseases.
Acknowledgements The work was supported by the National Natural Science Foundation of China
(31520103908, 31970905), Hebei Provincial Natural Science Foundation (C2017205140), Hebei
Provincial Education Department Foundation of China (ZD2015105), Hebei Provincial Fund for
studying abroad and returning to China (C201862), and Hebei normal University Outstanding Youth
Fund (L2018J05).
40 P. Yu and Y.-Z. Chang
References
21. Faucheux BA, Nillesse N, Damier P, Spik G, Mouatt-Prigent A, Pierce A, Leveugle B, Kubis
N, Hauw JJ, Agid Y et al (1995) Expression of lactoferrin receptors is increased in the mesen-
cephalon of patients with Parkinson disease. Proc Natl Acad Sci USA 92(21):9603–9607
22. Fillebeen C, Dehouck B, Benaissa M, Dhennin-Duthille I, Cecchelli R, Pierce A (1999) Tumor
necrosis factor-alpha increases lactoferrin transcytosis through the blood-brain barrier. J Neu-
rochem 73(6):2491–2500
23. Fillebeen C, Ruchoux MM, Mitchell V, Vincent S, Benaissa M, Pierce A (2001) Lactoferrin
is synthesized by activated microglia in the human substantia nigra and its synthesis by the
human microglial CHME cell line is upregulated by tumor necrosis factor alpha or 1-methyl-
4-phenylpyridinium treatment. Brain Res Mol Brain Res 96(1–2):103–113
24. Fisher J, Devraj K, Ingram J, Slagle-Webb B, Madhankumar AB, Liu X, Klinger M, Simpson
IA, Connor JR (2007) Ferritin: a novel mechanism for delivery of iron to the brain and other
organs. Am J Physiol Cell Physiol 293(2):C641–C649
25. Galy B, Ferring D, Benesova M, Benes V, Hentze MW (2004) Targeted mutagenesis of the
murine IRP1 and IRP2 genes reveals context-dependent RNA processing differences in vivo.
RNA (New York, NY) 10(7):1019–1025
26. Ganz T, Nemeth E (2012) Hepcidin and iron homeostasis. Biochimica et biophysica acta
1823(9):1434–1443
27. Gao G, You L-H, Chang Y-Z (2017) Iron metabolism in Parkinson’s disease. In: Oxidative
stress and redox signalling in Parkinson’s disease. The Royal Society of Chemistry, pp 255–276
(Chapter 9)
28. Ghosh MC, Zhang DL, Rouault TA (2015) Iron misregulation and neurodegenerative disease
in mouse models that lack iron regulatory proteins. Neurobiol Dis 81:66–75
29. Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF, Boron WF, Nussberger S,
Gollan JL, Hediger MA (1997) Cloning and characterization of a mammalian proton-coupled
metal-ion transporter. Nature 388(6641):482–488
30. Han J, Day JR, Connor JR, Beard JL (2003) Gene expression of transferrin and transferrin
receptor in brains of control vs. iron-deficient rats. Nutr Neurosci 6(1):1–10
31. Healy S, McMahon J, Owens P, FitzGerald U (2016) Significant glial alterations in response
to iron loading in a novel organotypic hippocampal slice culture model. Sci Rep 6:36410
32. Healy S, McMahon JM, FitzGerald U (2017) Modelling iron mismanagement in neurode-
generative disease in vitro: paradigms, pitfalls, possibilities & practical considerations. Prog
Neurobiol 158:1–14
33. Hentze MW, Muckenthaler MU, Andrews NC (2004) Balancing acts: molecular control of
mammalian iron metabolism. Cell 117(3):285–297
34. Hentze MW, Muckenthaler MU, Galy B, Camaschella C (2010) Two to tango: regulation of
Mammalian iron metabolism. Cell 142(1):24–38
35. Jeong SY, David S (2003) Glycosylphosphatidylinositol-anchored ceruloplasmin is required
for iron efflux from cells in the central nervous system. J Biol Chem 278(29):27144–27148
36. Kautz L, Jung G, Du X, Gabayan V, Chapman J, Nasoff M, Nemeth E, Ganz T (2015) Ery-
throferrone contributes to hepcidin suppression and iron overload in a mouse model of beta-
thalassemia. Blood 126(17):2031–2037
37. Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T (2014) Identification of erythroferrone
as an erythroid regulator of iron metabolism. Nat Genet 46(7):678–684
38. Ke Y, Qian ZM (2003) Iron misregulation in the brain: a primary cause of neurodegenerative
disorders. Lancet Neurol 2(4):246–253
39. Ke Y, Qian ZM (2007) Brain iron metabolism: neurobiology and neurochemistry. Prog Neu-
robiol 83(3):149–173
40. Kim HY, LaVaute T, Iwai K, Klausner RD, Rouault TA (1996) Identification of a conserved and
functional iron-responsive element in the 5 -untranslated region of mammalian mitochondrial
aconitase. J Biol Chem 271(39):24226–24230
41. Klomp LW, Farhangrazi ZS, Dugan LL, Gitlin JD (1996) Ceruloplasmin gene expression in
the murine central nervous system. J Clin Investig 98(1):207–215
42 P. Yu and Y.-Z. Chang
42. Klomp LW, Gitlin JD (1996) Expression of the ceruloplasmin gene in the human retina
and brain: implications for a pathogenic model in aceruloplasminemia. Hum Mol Genet
5(12):1989–1996
43. Krause A, Neitz S, Magert HJ, Schulz A, Forssmann WG, Schulz-Knappe P, Adermann K
(2000) LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity.
FEBS Lett 480(2–3):147–150
44. Lai CH, Kuo KH (2005) The critical component to establish in vitro BBB model: Pericyte.
Brain Res Brain Res Rev 50(2):258–265
45. Lane DJ, Robinson SR, Czerwinska H, Bishop GM, Lawen A (2010) Two routes of iron
accumulation in astrocytes: ascorbate-dependent ferrous iron uptake via the divalent metal
transporter (DMT1) plus an independent route for ferric iron. Biochem J 432(1):123–132
46. LaVaute T, Smith S, Cooperman S, Iwai K, Land W, Meyron-Holtz E, Drake SK, Miller G,
Abu-Asab M, Tsokos M, Switzer R 3rd, Grinberg A, Love P, Tresser N, Rouault TA (2001)
Targeted deletion of the gene encoding iron regulatory protein-2 causes misregulation of iron
metabolism and neurodegenerative disease in mice. Nat Genet 27(2):209–214
47. Li H, Sun H, Qian ZM (2002) The role of the transferrin-transferrin-receptor system in drug
delivery and targeting. Trends Pharmacol Sci 23(5):206–209
48. Ma J, Zhang FL, Zhou G, Bao YX, Shen Y, Qian ZM (2018) Different characteristics of hepcidin
expression in IL-6+/+ and IL-6-/- neurons and astrocytes treated with lipopolysaccharides.
Neurochem Res 43(8):1624–1630
49. Malecki EA, Devenyi AG, Beard JL, Connor JR (1999) Existing and emerging mechanisms
for transport of iron and manganese to the brain. J Neurosci Res 56(2):113–122
50. McCarthy RC, Kosman DJ (2013) Ferroportin and exocytoplasmic ferroxidase activity are
required for brain microvascular endothelial cell iron efflux. J Biol Chem 288(24):17932–17940
51. McCarthy RC, Kosman DJ (2014) Glial cell ceruloplasmin and hepcidin differentially regulate
iron efflux from brain microvascular endothelial cells. PLoS ONE 9(2):e89003
52. McCarthy RC, Kosman DJ (2015) Mechanisms and regulation of iron trafficking across the
capillary endothelial cells of the blood-brain barrier. Front Mol Neurosci 8:31
53. McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, Sager G, Mudaly E, Mudaly M, Richardson
C, Barlow D, Bomford A, Peters TJ, Raja KB, Shirali S, Hediger MA, Farzaneh F, Simpson
RJ (2001) An iron-regulated ferric reductase associated with the absorption of dietary iron.
Science (New York, NY) 291(5509):1755–1759
54. Meyron-Holtz EG, Ghosh MC, Iwai K, LaVaute T, Brazzolotto X, Berger UV, Land W,
Ollivierre-Wilson H, Grinberg A, Love P, Rouault TA (2004) Genetic ablations of iron regula-
tory proteins 1 and 2 reveal why iron regulatory protein 2 dominates iron homeostasis. EMBO
J 23(2):386–395
55. Moos T (2002) Brain iron homeostasis. Dan Med Bull 49(4):279–301
56. Moos T, Morgan EH (2000) Transferrin and transferrin receptor function in brain barrier sys-
tems. Cell Mol Neurobiol 20(1):77–95
57. Moos T, Rosengren Nielsen T, Skjorringe T, Morgan EH (2007) Iron trafficking inside the
brain. J Neurochem 103(5):1730–1740
58. Moos T, Trinder D, Morgan EH (2000) Cellular distribution of ferric iron, ferritin, transfer-
rin and divalent metal transporter 1 (DMT1) in substantia nigra and basal ganglia of nor-
mal and beta2-microglobulin deficient mouse brain. Cell Mol Biol (Noisy-le-Grand, France)
46(3):549–561
59. Moroo I, Ujiie M, Walker BL, Tiong JW, Vitalis TZ, Karkan D, Gabathuler R, Moise AR,
Jefferies WA (2003) Identification of a novel route of iron transcytosis across the mammalian
blood-brain barrier. Microcirculation (New York, NY: 1994) 10(6):457–462
60. Morris CM, Candy JM, Oakley AE, Bloxham CA, Edwardson JA (1992) Histochemical dis-
tribution of non-haem iron in the human brain. Acta Anat 144(3):235–257
61. Muckenthaler MU, Galy B, Hentze MW (2008) Systemic iron homeostasis and the iron-
responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr
28:197–213
3 Brain Iron Metabolism and Regulation 43
62. Nemeth E, Ganz T (2006) Regulation of iron metabolism by hepcidin. Annu Rev Nutr
26:323–342
63. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan J (2004)
Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization.
Science (New York, NY) 306(5704):2090–2093
64. Oshiro S, Kawahara M, Kuroda Y, Zhang C, Cai Y, Kitajima S, Shirao M (2000) Glial cells
contribute more to iron and aluminum accumulation but are more resistant to oxidative stress
than neuronal cells. Biochim Biophys Acta 1502(3):405–414
65. Oshiro S, Morioka MS, Kikuchi M (2011) Dysregulation of iron metabolism in Alzheimer’s dis-
ease, Parkinson’s disease, and amyotrophic lateral sclerosis. Adv Pharmacol Sci 2011:378278
66. Pantopoulos K (2004) Iron metabolism and the IRE/IRP regulatory system: an update. Ann N
Y Acad Sci 1012:1–13
67. Patel BN, David S (1997) A novel glycosylphosphatidylinositol-anchored form of ceruloplas-
min is expressed by mammalian astrocytes. J Biol Chem 272(32):20185–20190
68. Patel BN, Dunn RJ, David S (2000) Alternative RNA splicing generates a
glycosylphosphatidylinositol-anchored form of ceruloplasmin in mammalian brain. J Biol
Chem 275(6):4305–4310
69. Pelizzoni I, Zacchetti D, Campanella A, Grohovaz F, Codazzi F (2013) Iron uptake in quiescent
and inflammation-activated astrocytes: a potentially neuroprotective control of iron burden.
Biochim Biophys Acta 1832(8):1326–1333
70. Peters DG, Connor JR, Meadowcroft MD (2015) The relationship between iron dyshomeosta-
sis and amyloidogenesis in Alzheimer’s disease: two sides of the same coin. Neurobiol Dis
81:49–65
71. Qian ZM, Chang YZ, Leung G, Du JR, Zhu L, Wang Q, Niu L, Xu YJ, Yang L, Ho KP, Ke Y
(2007) Expression of ferroportin1, hephaestin and ceruloplasmin in rat heart. Biochem Biophys
Acta 1772(5):527–532
72. Qian ZM, He X, Liang T, Wu KC, Yan YC, Lu LN, Yang G, Luo QQ, Yung WH, Ke Y
(2014) Lipopolysaccharides upregulate hepcidin in neuron via microglia and the IL-6/STAT3
signaling pathway. Mol Neurobiol 50(3):811–820
73. Qian ZM, Pu YM, Wang Q, Ke Y, Yao YD, Chen WF, Shen X, Feng YM, Tang PL (1999) Cere-
bellar granule cells acquire transferrin-free iron by a carrier-mediated process. Neuroscience
92(2):577–582
74. Qian ZM, Tang PL, Wang Q (1997) Iron crosses the endosomal membrane by a carrier-mediated
process. Prog Biophys Mol Biol 67(1):1–15
75. Ramey G, Deschemin JC, Durel B, Canonne-Hergaux F, Nicolas G, Vaulont S (2010) Hepcidin
targets ferroportin for degradation in hepatocytes. Haematologica 95(3):501–504
76. Rasmussen MK, Mestre H, Nedergaard M (2018) The glymphatic pathway in neurological
disorders. Lancet Neurol 17(11):1016–1024
77. Rouault TA (2013) Iron metabolism in the CNS: implications for neurodegenerative diseases.
Nat Rev Neurosci 14(8):551–564
78. Rouault TA, Zhang DL, Jeong SY (2009) Brain iron homeostasis, the choroid plexus, and
localization of iron transport proteins. Metab Brain Dis 24(4):673–684
79. Ryan F, Zarruk JG, Losslein L, David S (2018) Ceruloplasmin plays a neuroprotective role in
cerebral ischemia. Frontiers in neuroscience 12:988
80. Siddappa AJ, Rao RB, Wobken JD, Leibold EA, Connor JR, Georgieff MK (2002) Develop-
mental changes in the expression of iron regulatory proteins and iron transport proteins in the
perinatal rat brain. J Neurosci Res 68(6):761–775
81. Skjorringe T, Moller LB, Moos T (2012) Impairment of interrelated iron- and copper home-
ostatic mechanisms in brain contributes to the pathogenesis of neurodegenerative disorders.
Front Pharmacol 3:169
82. Smith SR, Ghosh MC, Ollivierre-Wilson H, Hang Tong W, Rouault TA (2006) Complete loss
of iron regulatory proteins 1 and 2 prevents viability of murine zygotes beyond the blastocyst
stage of embryonic development. Blood Cells Mol Dis 36(2):283–287
44 P. Yu and Y.-Z. Chang
83. Song N, Wang J, Jiang H, Xie J (2018) Astroglial and microglial contributions to iron
metabolism disturbance in Parkinson’s disease. Biochimica et biophysica acta 1864(3):967–973
84. Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP, Arredondo M, Gonzalez-Billault C, Nunez
MT (2013) Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes
iron accumulation in central nervous system cells. J Neurochem 126(4):541–549
85. Vargas JD, Herpers B, McKie AT, Gledhill S, McDonnell J, van den Heuvel M, Davies KE,
Ponting CP (2003) Stromal cell-derived receptor 2 and cytochrome b561 are functional ferric
reductases. Biochem Biophys Acta 1651(1–2):116–123
86. Wang J, Jiang H, Xie JX (2007) Ferroportin1 and hephaestin are involved in the nigral iron
accumulation of 6-OHDA-lesioned rats. Eur J Neurosci 25(9):2766–2772
87. Wang L, Liu X, You LH, Ci YZ, Chang S, Yu P, Gao G, Chang YZ (2019) Hepcidin and iron
regulatory proteins coordinately regulate ferroportin 1 expression in the brain of mice. J Cell
Physiol 234(5):7600–7607
88. Wang SM, Fu LJ, Duan XL, Crooks DR, Yu P, Qian ZM, Di XJ, Li J, Rouault TA, Chang
YZ (2010) Role of hepcidin in murine brain iron metabolism. Cell Mol Life Sci CMLS
67(1):123–133
89. Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L (2014) The role of iron in brain ageing
and neurodegenerative disorders. Lancet Neurol 13(10):1045–1060
90. Wu LJ, Leenders AG, Cooperman S, Meyron-Holtz E, Smith S, Land W, Tsai RY, Berger
UV, Sheng ZH, Rouault TA (2004) Expression of the iron transporter ferroportin in synaptic
vesicles and the blood-brain barrier. Brain Res 1001(1–2):108–117
91. Xu H, Wang Y, Song N, Wang J, Jiang H, Xie J (2017) New progress on the role of glia in
iron metabolism and iron-induced degeneration of dopamine neurons in Parkinson’s disease.
Frontiers in molecular neuroscience 10:455
92. Yang WM, Jung KJ, Lee MO, Lee YS, Lee YH, Nakagawa S, Niwa M, Cho SS, Kim DW
(2011) Transient expression of iron transport proteins in the capillary of the developing rat
brain. Cell Mol Neurobiol 31(1):93–99
93. Yu P, Qian ZM, Duan XL, Chang YZ (2004) The structure, gene expression of Divalent Metal
Transporter 1 and the underlying regulatory mechanisms. Chin J Neuroanat 20:205–209
94. Zanardi A, Conti A, Cremonesi M, D’Adamo P, Gilberti E, Apostoli P, Cannistraci CV, Piperno
A, David S, Alessio M (2018) Ceruloplasmin replacement therapy ameliorates neurological
symptoms in a preclinical model of aceruloplasminemia. EMBO Mol Med 10(1):91–106
95. Zechel S, Huber-Wittmer K, von Bohlen und Halbach O (2006) Distribution of the iron-
regulating protein hepcidin in the murine central nervous system. J Neurosci Res 84(4):790–800
96. Zhang P, Land W, Lee S, Juliani J, Lefman J, Smith SR, Germain D, Kessel M, Leapman R,
Rouault TA, Subramaniam S (2005) Electron tomography of degenerating neurons in mice
with abnormal regulation of iron metabolism. J Struct Biol 150(2):144–153
97. Zheng W, Monnot AD (2012) Regulation of brain iron and copper homeostasis by brain barrier
systems: implication in neurodegenerative diseases. Pharmacol Ther 133(2):177–188
98. Zumbrennen-Bullough KB, Becker L, Garrett L, Holter SM, Calzada-Wack J, Mossbrugger
I, Quintanilla-Fend L, Racz I, Rathkolb B, Klopstock T, Wurst W, Zimmer A, Wolf E, Fuchs
H, Gailus-Durner V, de Angelis MH, Romney SJ, Leibold EA (2014) Abnormal brain iron
metabolism in Irp2 deficient mice is associated with mild neurological and behavioral impair-
ments. PLoS ONE 9(6):e98072
Chapter 4
Iron Pathophysiology in Parkinson
Diseases
Hong Jiang, Ning Song, Qian Jiao, Limin Shi and Xixun Du
Abstract The key molecular events that provoke Parkinson’s disease (PD) are not
fully understood. Iron deposit was found in the substantia nigra pars compacta
(SNpc) of PD patients and animal models, where dopaminergic neurons degeneration
occurred selectively. The mechanisms involved in disturbed iron metabolism remain
unknown, however, considerable evidence indicates that iron transporters dysregula-
tion, activation of L-type voltage-gated calcium channel (LTCC) and ATP-sensitive
potassium (KATP) channels, as well as N-methyl-D-aspartate (NMDA) receptors
(NMDARs) contribute to this process. There is emerging evidence on the structural
links and functional modulations between iron and α-synuclein, and the key player
in PD which aggregates in Lewy bodies. Iron is believed to modulate α-synuclein
synthesis, post-translational modification, and aggregation. Furthermore, glia, espe-
cially activated astroglia and microglia, are involved in iron deposit in PD. Glial
contributions were largely dependent on the factors they released, e.g., neurotrophic
factors, pro-inflammatory factors, lactoferrin, and those undetermined. Therefore,
iron chelation using iron chelators, the extracts from many natural foods with iron
chelating properties, may be an effective therapy for prevention and treatment of the
disease.
tion, and other dysautonomic symptoms [4–6]. Up to now, the key molecular events
that provoke neurodegeneration are not fully understood. In addition to misfolding
of proteins, impairment of the ubiquitin proteasome, dysfunction of mitochondria,
oxidative stress, there is mounting evidence implicated that iron accumulation is
pivotal to PD pathogenesis [7, 8].
In the late 1980s, several groups reported elevated iron levels in the SN of parkin-
sonian patients compared to age-matched control [9–12]. In other parts of brain,
including cortex (Brodmann area 21), hippocampus, putamen, and globus pallidus,
the contents of iron were unchanged [9]. Especially, it was reported that SNpc is a
selective area of iron accumulation and the ratio of ferrous to ferric iron decreased
in parkinsonian patients [13]. Furthermore, a variety of methods, such as inductively
coupled plasma spectroscopy, magnetic resonance imaging, laser microprobe mass
analysis, susceptibility-weighted imaging, and enhanced T2-star-weighted angiog-
raphy, have proved the increased iron levels in the SNpc of PD brains [7]. The
disruption of iron metabolism in parkinsonian patients is also demonstrated by a
previous work that the iron levels are significantly decreased in the temporal cor-
tex in brain when compared with age-matched healthy controls, suggesting a likely
re-distribution pattern in PD [14].
The iron metabolism disturbance was also present in PD animal and cell models.
The transgenic PD mice model overexpressing the mutant human A53T α-synuclein
(A53T mice) show an accumulation of aggregates comprising α-synuclein and
develop age-dependent motor deficits. A53T mice are more vulnerable to accumulate
iron in the SN than the wild type [15]. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
(MPTP), an inhibitor of complex I of the mitochondrial electron transport chain, is
commonly used to induce PD animal models that exhibit the damage of dopaminergic
neurons [1, 16]. Injection of MPTP into the caudate or putamen of monkeys resulted
in the increase of ferric iron-reaction products in the ipsilateral SNpc [17, 18]. In
hemiparkinsonian monkeys, iron contents were significantly increased from 4.5 to
18 months in the ipsilateral SNpc after MPTP injection by unilateral internal carotid
artery [19]. Moreover, the increasing extent was significantly correlated to the extent
of dopaminergic cell death [17, 19, 20]. Additionally, the phenomena that nigral
iron levels elevation is also observed in MPTP-induced PD mice model [21–23].
6-hydroxydopamine (6-OHDA) is a neurotoxin, which induces dopaminergic neu-
rons death and therefore has been implicated in PD rat models. It was reported that
iron levels increased in the ipsilateral SN from 1 to 14 days in 6-OHDA unilaterally
lesioned rats [24]. Actually, both increased iron levels and dopaminergic neurons
loss are apparent in the SN as promptly as one day following 6-OHDA injection
[24]. Iron levels increased in SN with 6-OHDA injection into rats have been proved
by peer laboratories [21, 25–27]. In vitro studies, iron uptake significantly increased
both in 1-methyl-4-phenylpyridinium (MPP+ )- and 6-OHDA-induced PD cell mod-
els [28, 29]. In other models of PD, including lipopolysaccharide-induced inflam-
mation models, the loss of dopaminergic neurons was accompanied by increased
iron and ferritin levels in glial cells of the SN pars reticulate [30, 31]. Moreover, in
iron-overloaded animal models, such as direct injection of ferric iron into the SN,
high dietary iron supplements and peripheral iron overload, they develop degener-
4 Iron Pathophysiology in Parkinson Diseases 47
ation of dopaminergic neurons and further illustrated the relationship between iron
and PD pathogenesis [15, 32]. With the common focusing on neurotoxins MPTP
and 6-OHDA, the environment toxins exiting in food and drinking water, including
paraquat, rotenone, and mancozeb, may be directly or indirectly involved and affect
iron metabolism [33, 34].
The unbalanced distribution of iron in the brain of parkinsonian patients indicates
that iron plays a key role in the degeneration of dopaminergic neurons. Potential
mechanisms have been offered to explain why iron metabolism disturbance occurs
(see below), as well as how elevated iron behaviors leading to dopaminergic neurons
degeneration [7]. Among these, iron and DA toxic couple has to be highlighted. DA
is the central neurotransmitter involved in PD. DA easily forms toxic metabolites
and these processes occur predominantly in the cytoplasm [35]. Physiologically,
H2 O2 is produced by a DA enzymatic process via monoamine oxidase. The revealed
aspects of iron-DA coupling are largely derived from their pro-oxidant properties
[36]. The high-level iron in dopaminergic neurons aggravates the oxidative stress
by Fenton reaction that iron reacts with H2 O2 produced in the enzymatic process
in DA metabolism and then can generate OH that can damage proteins, nucleic
acids, and membrane phospholipids [37]. An alternative mechanism has been raised
that the oxidation of DA by iron forms 6-OHDA. As a commonly used neurotoxin
in PD models, 6-OHDA even liberates iron from ferritin and produced H2 O2 in
its metabolism. Recently, it was reported that high-level iron inside cells caused
ferroptosis which is a form of regulated cell death characterized by the iron-dependent
accumulation of lipid hydroperoxides to lethal levels [38, 39]. Elevated iron likely
contributes in dopaminergic neurons degeneration in PD, which could be further
validated by the facts that using pharmacological or genetic chelation of iron in
animal models of PD has neuroprotective effects on dopaminergic neurons (see
below) [40, 41].
TfR, namely TfR1 and TfR2. TfR1, expressed abundantly in the central nervous
system, especially in neurons, constitutes the main receptor for Tf and internalizes
it through receptor-mediated endocytosis. Furthermore, TfR1 mRNA contains iron-
responsive elements (IRE) in the 5 -untranslated region, thus it can be regulated
by intracellular iron levels via iron response proteins (IRP) [42]. In contrast, TfR2,
mainly expressed in the mitochondria of nigral dopaminergic neurons [43], does not
contain the IRE, thus it cannot be regulated by iron levels. Although TfR2 has a lower
affinity for Tf than TfR1, it can serve as an iron sensor in the regulation of hepcidin
[44]. Recent studies found that 1-methyl-4-phenylpyridinium (MPP+ ) treatment led
to the increased expression of TfR, suggesting a role for TfR-dependent iron influx
in the selective iron accumulation in PD [45]. The localization of TfR2 in the mito-
chondria of dopaminergic neurons and a dramatic increase of oxidized Tf in the SN
raised the possibility that the Tf-TfR2 system might also contribute to iron deposit
in PD [46].
The non-transferrin-bound iron (NTBI) is mainly transported through divalent
metal transporter 1 (DMT1), which is responsible for ferrous iron uptake. There
are at least four distinct DMT1 mRNA isoforms: 1A, 1B, +IRE and −IRE [47, 48].
DMT1, also known as natural resistance-associated macrophage protein 2 (Nramp 2)
or divalent cation transporter 1 (DCT1), was first discovered to transport iron in
the duodenal lumen. In the SN, DMT1 is widely expressed in neurons, astrocytes
and microglia [49], which mediates iron transmembrane transport as well as from
the endosomes to the cytoplasm [50]. The expression of DMT1 can be regulated
post-transcriptionally through IRE/IRP, or post-translationally through the ubiquitin-
proteasome system (UPS). Previous studies have shown that the E3 ligase parkin is
responsible for the proteasomal degradation of DMT1 and regulates iron transport
[51]. Moreover, DMT1 can be ubiquitinated by the neural precursor cell expressed
developmentally down-regulated protein 4 (Nedd4) family member WW domain-
containing protein 2 (WWP2), and this requires Nedd4 family interacting protein
1 (Ndfip1) and/or Ndfip2 [52]. In the postmortem brains of PD patients and ani-
mal models, the expression of nigral DMT1 was significantly up-regulated, thus
contributed to the intracellular iron accumulation and dopaminergic neurons degen-
eration [28, 53, 54]. Modulation of DMT1 proteasomal degradation via UPS might
also be associated with DMT1 dysregulation and participate in iron deposit in PD
[51, 55, 56].
Another iron uptake mechanism involves an iron transporter called lactoferrin
(Lf), which acts as an iron-binding protein, belonging to the Tf family. In a similar
manner to Tf, Lf-bound ferric iron can bind to Lf receptor (LfR), resulting in iron
transport across the plasma membrane. The affinity of Lf for iron is 300 times higher
than that of Tf [57]. In the brain, Lf is only synthesized and released by activated
microglia [58]. LfR is present in blood vessels and nigral dopaminergic neurons
[59], suggesting that Lf-LfR pathway may be involved in iron transport into brain
parenchyma and dopaminergic neurons. Moreover, an increased expression of Lf
and LfR on dopaminergic neurons was found in PD patients [58, 59], which may
account for the excessive accumulation of iron. And this increase was occurred in
4 Iron Pathophysiology in Parkinson Diseases 49
NMDARs are cation channels which mediate Na+ and Ca2+ ions entering the cells and
could be activated by glutamate, glycine, or D-serineare. NMDARs activation appro-
priately plays a critical role in several physiological processes, e.g., synaptic plas-
ticity and excitatory neurotransmission. However, excessive activation of NMDARs
contributes to pathological changes in the brain. NMDARs mediated excessive Ca2+
influx might induce neuronal death in several neurodegenerative diseases, including
PD [71, 72]. NR1, a very important subunit of NMDARs, which expression was
increased in surviving dopaminergic neurons from PD brains compared with the
controls [73]. Some studies also revealed that NMDARs activation could signifi-
cantly promote Fe2+ entry into cells. The underlying mechanisms might be involved.
First, NMDARs activation increases NTBI influx. Fe2+ could compete with Ca2+ for
NMDARs to enter the cultured neurons [74, 75]. The increased iron levels may induce
a corresponding ROS overproduction and higher susceptibility of neurons. Another
investigation has identified a novel signaling cascade for NMDARs regulated iron
uptake in brain [76]. It was reported that glutamate via NMDARs trigger Ca2+ influx,
then activated neuronal nitric oxide (NO) synthase (NOS) produce NO, which induce
excitability toxicity [76–78]. And neuronal NOS (nNOS) binding to CAPON could
cause NO delivery to Dexrase1 and S-nitrosylation of Dexrase1, which promote iron
uptake by regulating the function of DMT1 [79]. Rhes (Dexras2), a homolog of
Dexras1, is selectively localized to the corpus striatum. Rhes was also reported to
be involved in NMDARs-induced iron influx into the striatal neurons and regulating
striatal iron homeostasis by PKA-Rhes-DMT1 pathway [80].
Second, NMDARs activation enhanced iron releasing from lysosome. The iron
in lysosome serves as a main source for intracellular iron, in which DMT1 plays a
critical role in iron recycling from lysosome to cytoplasm. Recently, several evidence
showed that Dexras1/PAP7/DMT1 complex located on the lysosome. Either impaired
DMT1 function or the collapsed proton gradient could reduce iron pool in cytoplasm,
which indicated that Dexras1/PAP7/DMT1 complex plays a role in iron release from
lysosome to cytoplasm [81].
Finally, NMDARs activation increased DMT1 expression. It has been reported
that the mRNA levels of DMT1 were increased after 5 min treated with 50 μM
NMDA to primary hippocampal cultures. With exposure to the transcription inhibitor
actinomycin D and NMDARs antagonist MK-801, DMT1 up-regulation induced by
NMDAs or 6-OHDA could be restored [82]. Interestingly, both excessive activation
of NMDAs and up-regulation of DMT1 were observed in the process of degeneration
of dopaminergic neurons in PD, indicating the close links between NMDARs and
DMT1 in PD [83].
4 Iron Pathophysiology in Parkinson Diseases 51
Functional KATP channels are composed of four pore-forming Kir6 (Kir6.1 and
Kir6.2) subunits and four regulatory SUR (SUR1, SUR2A, and SUR2B) subunits.
Different subunit compositions decided the different biophysical, pharmacological,
and metabolic properties of KATP channels [84]. In dopaminergic neurons in the SN
and VTA, KATP channels are both expressed with Kir6.2 and SUR1 subunits. But
only the KATP channels of dopaminergic neurons in the SN were selectively activated
when treated with MPP+ . And dopaminergic neurons in the SN but not those in the
VTA in the MPTP model were selectively rescued after genetic inactivation of Kir6.2
subunits [85]. It was also reported that mRNA levels of the regulatory subunit SUR1
were increased in survived dopaminergic neurons in the SN from PD patients. Other-
wise, mRNA levels of SUR2 and Kir6.2 were not changed [73]. These results suggest
that the activation of KATP channels might be involved in the selective degeneration
of dopaminergic neurons in the SN in PD. The activation of KATP channels would
induce the membrane potential hyperpolarized, which is important for the membrane
oscillations that underlie bursting firing generated [86]. KATP channels’ gated burst
firing could aggravate excitotoxicity and increase calcium loading with NMDARs
and LTCCs synergistically in the dopaminergic neurons of SN, where metabolic
state have already challenged [87]. This led to the insufficient mitochondrial calcium
buffering capacities and acceleration of calcium-induced ROS production [88], which
in turn activated KATP channels in highly vulnerable neurons [73].
Selective activation of KATP channels in the SN was suggested to contribute to
iron accumulation. The transport function of DMT1 is proton-coupled and dependent
on the cell membrane potential. It was reported that hyperpolarized potentials could
promote the iron uptake via DMT1 [89]. There is evidence that diazoxide, a KATP
channel opener, could increase intracellular iron levels after ferrous iron treatment
[90]. The subsequent ATP depletion and ROS production would induce additional
KATP channels activated in a feed-forward cycle [73]. Accordingly, inhibition of
KATP channels dramatically decreases the iron uptake and inhibits cell damage [90].
These provide evidence that the selective iron accumulation in the SN might be
related to KATP channels activation; further investigation should be done to reveal
the underlying mechanisms.
indicated an increased nigral iron level. Thus, hyperechogenicity has been partially
shown to be increased iron levels in the SN [92], which has also been deemed to be
a typical sign in idiopathic PD. A subset of cellular proteins not degraded attributed
to Parkin mutations are currently supposed to be the most common reasons of famil-
ial Parkinsonism [93–95]. Iron could induce altered Parkin solubility and result in
its intracellular aggregation. And with the depletion of soluble, functional forms of
Parkin, proteasomal activities were impaired with cell damage [96]. In 1998, Parkin
mutation was first identified in autosomal recessive juvenile parkinsonism (ARJP),
and iron staining in the SN was reported to be more intense than that of controls, as
well as sporadic PD patients [91]. It was hypothesized that iron accumulation might
be related to the loss of the Parkin gene.
More recently, Parkin was reported to be responsible for ubiquitination of
DMT1 + IRE. The expression of 1B-DMT1 isoforms was decreased if SH-SY5Y
cells overexpressed with Parkin [51]. When fed with iron-supplemented diet, trans-
genic mice with DMT1 overexpression showed selectively accumulated iron in the
SN, meanwhile, the expression of Parkin is also up-regulated, likely a neuroprotec-
tive response [97]. In human lymphocytes containing a homozygous deletion of exon
4 of Parkin or in the brains of Parkin knockout animals, expression of DMT1 + IRE
was also shown to be elevated.
Iron accumulation and oxidative stress induced by ferrous ammonium was able to
up-regulate α-synuclein protein levels in these cells. The data showed the 5 -UTR
in α-synuclein is a positive mediator of promoting translation, but not of mRNA
stability/expression, consistent with the fact that IRPs serve to block transcription
activation [104]. On the other hand, in HEK293 cells iron chelator deferoxamine
(DFO, also known as desferal) decreased human α-synuclein mRNA levels [105].
Together with the evidence of an IRE presence in the 5 -UTR of the Alzheimer’s
amyloid precursor protein (APP) transcript [106], these evidence suggests that iron
might contribute to neurodegenerative disease course by up-regulating α-synuclein
or APP levels.
Alpha-synuclein undergoes a variety of post-translational modifications, which
have been linked to the aggregation and pathology of α-synuclein [107, 108]. Nitra-
tion and phosphorylation were believed to be associated with iron and iron-induced
oxidative stress. Nitrated α-synuclein is present in LBs, as well as in the insoluble
fractions of affected brain regions in PD [109]. It is suggested that nitration of only
a single tyrosine residue is sufficient to induce profound changes in catalytic activ-
ities and structural conformations of proteins. The attachment of a nitro molecule
to α-synuclein at tyrosine residues (Y39, Y125, Y133, and Y136), is sufficient to
induce profound changes in α-synuclein [110]. Nitrated of α-synuclein monomers
and dimers could be mixed into the fibrils and accelerate the rate of fibril formation
of unmodified α-synuclein [111, 112]. It was demonstrated that nitrated monomeric
α-synuclein was degraded at a slower rate; together with diminished binding to
lipid vesicles, nitrated α-synuclein were responsible for increasing fibrils formation
and intracytoplasmic inclusions [113]. Excessive iron levels are associated with the
increase of oxidative/nitrative stress, leading to elevated levels of tyrosine nitration.
Iron supplements in rats markedly increased the extent of lipid peroxidation, protein
carbonyls tyrosine nitration and oxidative metabolism of NO in liver [114]. There are
currently no reports on the effects of iron on α-synuclein nitration. However, oxida-
tive stress, the most common deleterious condition induced by iron, was believed to
be involved in the nitrated process of α-synuclein. In vitro evidence demonstrated that
microglial activation could induce NO-dependent oxidative stress in dopaminergic
neurons and then cause α-synuclein nitration. These nitrated, aggregated α-synuclein
during oxidative stress responses, in turn, incurs inflammatory microglial functions
[115, 116]. In vivo, nitrated-α-synuclein (Tyr125, Tyr133) levels was elevated in the
thymus of MPTP induced PD mice; nitrated recombinant α-synuclein was able to
activate microglia and astrocytes lead to the degeneration of dopaminergic neurons
[117, 118].
Phosphorylation of α-synuclein represents more than 90% of the total α-synuclein
found in LB, whereas only 4% of the soluble monomeric α-synuclein is phosphory-
lated physiologically [119]. Phosphorylation at S129 is the major post-translational
modification of α-synuclein [120]. Genetic approaches mimicking phosphorylated
α-synuclein at S129 was associated with pathology in a transgenic Drosophila model,
as well as in MPTP-treated monkeys. The mutation of serine 129 to aspartate signif-
icantly promotes α-synuclein phosphorylation and aggregation and thus dopaminer-
gic neurons death. However, altering serine 129 to alanine prevents phosphorylation
54 H. Jiang et al.
The role of iron in the pathology of PD has logically led to the theory that iron
chelation may be an effective therapy for prevention and treatment of the dis-
ease [4, 159–163]. Mice with low iron diets for six weeks may provide protec-
tion against MPTP-inducing insults [164]. Similarly, rats received an iron-deficient
diet attenuated kainate-induced neurotoxicity [165]. The clinically iron chelators,
DFO, deferiprone, and deferasirox, have been shown to attenuate 6-OHDA, MPTP
or iron-induced oxidative stress, mitochondrial dysfunction and prevent α-synuclein
aggregation both in vivo and in vitro [166–170]. Recent phase II clinical trial data
also demonstrated that deferiprone improved motor symptoms of PD patients [41].
8-hydroxyquinoline (8-HQ), a highly lipophilic complex, has been utilized as the
base for VK-28, M30, clioquinol, and Q1. These iron chelators readily penetrate cell
membranes and the blood-brain barrier and have been shown to be effective in both
cell culture and the PD animal models [40, 171–175].
Recently, several novel iron chelators have been explored and synthesized.
Lipophilic analogs of desferrioxamine B (conjugating ancillary compounds onto
4 Iron Pathophysiology in Parkinson Diseases 57
the amine terminus) attenuated neuronal loss in SK-N-BE2-M17 cell lines and
MPTP-treated mice [176, 177]. A novel hybrid iron chelator, (-)-12 (D-607), res-
cued PC12 cells from toxicity induced by iron [178]. Iron chelation therapeutic
nanoparticles protected by a zwitterionic poly(2-methacryloyloxyethyl phosphoryl-
choline) (PMPC), which delay the saturation of iron chelators in blood circula-
tion and prolong the in vivo lifetime, provided a PD phenotype reversion therapy
and significantly improved the living quality of the PD mice [179]. 7,8-dihydroxy-
4-((methylamino)methyl)-2H-chromen-2-one (DHC12) chelated mitochondrial and
cytoplasmic labile iron, protected mitochondria against lipid peroxidation, and antag-
onized MPTP-induced neuronal death [180]. Polysubstituted piperazine derivatives
(a selection of 1-hydroxypyridin-2-one (1,2-HOPO) metal chelators) and PBT434
(an orally bioavailable 8-hydroxyquinazolin-4(3H)-one) have also been designed as
new iron chelators and proved to be neuroprotective in dopaminergic cell lines and
multiple models of PD [181–183].
It has been found that the extracts from many natural foods have iron-chelating
properties. For example, epigallocatechin gallate (EGCG), a major polyphenol in
green tea, regulated the iron-export protein FPN1 and exerted a neurorescuing effect
in MPTP-induced PD mice and α-synuclein transfected-PC12 cells [184, 185].
Indeed, epidemiological evidence showed that drinking two cups of tea a day could
reduce the risk of PD [186]. Several natural flavonoids, such as ginkgetin (a nat-
ural bioflavonoid isolated from leaves of Ginkgo biloba L), curcumin (1,7-bis[4-
hydroxy 3-methoxy phenyl]-1,6-heptadiene-3,5-dione, the polyphenolic flavonoid
from Curcuma longa), myricetin and quercetin (bioflavonoids found in abundance
in fruits, vegetables, and herbs) exerted neuroprotective effects against neuroinjury
in PD model induced by MPTP or 6-OHDA via chelating iron [187–190]. Baicalin,
a major active ingredient of the plant Scutellaria baicalensis Georgi, has been shown
to reduce iron accumulation and have protective effects on dopaminergic neurons
in the SN of rotenone-induced PD rats, as well as in C6 cells [191, 192]. Ginseno-
sides, the principal active components of ginseng, also reduced nigral iron levels in
MPTP/MPP+ -treated models via regulating the expression of DMT1 and FPN1 [21,
193, 194]. A major advantage of these natural chelators is the mixture of neuropro-
tective properties they possess. In addition to iron chelation, they have been shown
to exert a variety of beneficial effects, including antioxidant, anti-inflammatory, anti-
apoptosis, and anticancer effects. Therefore, this allows for a more comprehensive
usage in the prevention and treatment of PD.
It is important to realize that whether and how iron chelators could remove excess
iron from the specified brain areas, without affecting systemic iron homeostasis.
Indeed, iron is an essential element in various physiological processes, e.g., DNA
synthesis, mitochondrial respiration, and oxygen transport. Future studies should
seek to identify more cellular specificity therapies to prevent and attenuate brain iron
metabolism dysfunction in PD.
58 H. Jiang et al.
References
25. He Y, Lee T, Leong SK (1999) Time course of dopaminergic cell death and changes in iron,
ferritin and transferrin levels in the rat substantia nigra after 6-hydroxydopamine (6-OHDA)
lesioning. Free Radic Res 31(2):103–112
26. Zhu Y et al (2017) Iron accumulation and microglia activation contribute to substantia nigra
hyperechogenicity in the 6-OHDA-induced rat model of Parkinson’s disease. Parkinsonism
Relat Disord 36:76–82
27. Olmedo-Diaz S et al (2017) An altered blood-brain barrier contributes to brain iron accumu-
lation and neuroinflammation in the 6-OHDA rat model of Parkinson’s disease. Neuroscience
362:141–151
28. Zhang S et al (2009) Up-regulation of divalent metal transporter 1 is involved in 1-
methyl-4-phenylpyridinium (MPP(+))-induced apoptosis in MES23.5 cells. Neurobiol Agin
30(9):1466–1476
29. Xu H et al (2010) Rg1 protects iron-induced neurotoxicity through antioxidant and iron
regulatory proteins in 6-OHDA-treated MES23.5 cells. J Cell Biochem 111(6):1537–1545
30. Zhang J et al (2005) Intrapallidal lipopolysaccharide injection increases iron and ferritin levels
in glia of the rat substantia nigra and induces locomotor deficits. Neuroscience 135(3):829–838
31. Zhang Z et al (2014) Pro-inflammatory cytokine-mediated ferroportin down-regulation con-
tributes to the nigral iron accumulation in lipopolysaccharide-induced Parkinsonian models.
Neuroscience 257:20–30
32. Ben-Shachar D, Youdim MB (1991) Intranigral iron injection induces behavioral and bio-
chemical “parkinsonism” in rats. J Neurochem 57(6):2133–2135
33. Nandipati S, Litvan I (2016) Environmental exposures and Parkinson’s disease. Int J Environ
Res Public Health 13(9)
34. Peng J, Oo ML, Andersen JK (2010) Synergistic effects of environmental risk factors and
gene mutations in Parkinson’s disease accelerate age-related neurodegeneration. J Neurochem
115(6):1363–1373
35. Xu J et al (2002) Dopamine-dependent neurotoxicity of α-synuclein: a mechanism for selective
neurodegeneration in Parkinson disease. Nat Med 8(6):600–606
36. Hare DJ, Double KL (2016) Iron and dopamine: a toxic couple. Brain 139:1026–1035
37. Dias V, Junn E, Mouradian MM (2013) The role of oxidative stress in Parkinson’s disease. J
Parkinsons Dis 3(4):461–491
38. Stockwell BR et al (2017) Ferroptosis: a regulated cell death nexus linking metabolism, redox
biology, and disease. Cell 171(2):273–285
39. Dixon SJ et al (2012) Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell
149(5):1060–1072
40. Kaur D et al (2003) Genetic or pharmacological iron chelation prevents MPTP-induced neu-
rotoxicity in vivo: a novel therapy for Parkinson’s disease. Neuron 37(6):899–909
41. Devos D et al (2014) Targeting chelatable iron as a therapeutic modality in Parkinson’s disease.
Antioxid Redox Signal 21(2):195–210
42. Trinder D, Baker E (2003) Transferrin receptor 2: a new molecule in iron metabolism. Int J
Biochem Cell Biol 35(3):292–296
43. Lee DW, Andersen JK (2010) Iron elevations in the aging Parkinsonian brain: a consequence
of impaired iron homeostasis? J Neurochem 112(2):332–339
44. Ward RJ et al (2014) The role of iron in brain ageing and neurodegenerative disorders. Lancet
Neurol 13(10):1045–1060
45. Kalivendi SV et al (2003) 1-Methyl-4-phenylpyridinium (MPP+ )-induced apoptosis and mito-
chondrial oxidant generation: role of transferrin-receptor-dependent iron and hydrogen per-
oxide. Biochem J 371(Pt 1):151–164
46. Mastroberardino PG et al (2009) A novel transferrin/TfR2-mediated mitochondrial iron trans-
port system is disrupted in Parkinson’s disease. Neurobiol Dis 34(3):417–431
47. Lee PL et al (1998) The human Nramp2 gene: characterization of the gene structure, alternative
splicing, promoter region and polymorphisms. Blood Cells Mol Dis 24(2):199–215
48. Hubert N, Hentze MW (2002) Previously uncharacterized isoforms of divalent metal trans-
porter (DMT)-1: implications for regulation and cellular function. Proc Natl Acad Sci U S A
99(19):12345–12350
60 H. Jiang et al.
99. Binolfi A et al (2006) Interaction of α-synuclein with divalent metal ions reveals key differ-
ences: a link between structure, binding specificity and fibrillation enhancement. J Am Chem
Soc 128(30):9893–9901
100. Uversky VN, Li J, Fink AL (2001) Metal-triggered structural transformations, aggregation,
and fibrillation of human α-synuclein. A possible molecular NK between Parkinson’s disease
and heavy metal exposure. J Biol Chem 276(47):44284–4496
101. Bharathi, Indi SS, Rao KS (2007) Copper- and iron-induced differential fibril formation in
α-synuclein: TEM study. Neurosci Lett 424(2):78–82
102. Friedlich AL, Tanzi RE, Rogers JT (2007) The 5’-untranslated region of Parkinson’s disease
α-synuclein messengerRNA contains a predicted iron responsive element. Mol Psychiatry
12(3):222–223
103. Li W et al (2011) Oxidative stress partially contributes to iron-induced α-synuclein aggregation
in SK-N-SH cells. Neurotox Res 19(3):435–442
104. Koukouraki P, Doxakis E (2016) Constitutive translation of human α-synuclein is mediated
by the 5’-untranslated region. Open Biol 6(4):160022
105. Febbraro F et al (2012) α-Synuclein expression is modulated at the translational level by iron.
NeuroReport 23(9):576–580
106. Rogers JT et al (2002) An iron-responsive element type II in the 5’-untranslated region of the
Alzheimer’s amyloid precursor protein transcript. J Biol Chem 277(47):45518–45528
107. Oueslati A, Fournier M, Lashuel HA (2010) Role of post-translational modifications in modu-
lating the structure, function and toxicity of α-synuclein: implications for Parkinson’s disease
pathogenesis and therapies. Prog Brain Res 183:115–145
108. Barrett PJ,. Timothy Greenamyre, J (2015) Post-translational modification of α-synuclein in
Parkinson’s disease. Brain Res 1628(Pt B):247–253
109. Giasson BI et al (2000) Oxidative damage linked to neurodegeneration by selective α-
synuclein nitration in synucleinopathy lesions. Science 290(5493):985–989
110. Reynolds MR, Berry RW, Binder LI (2007) Nitration in neurodegeneration: deciphering the
“Hows” “nYs”. Biochemistry 46(25):7325–7336
111. Burai R et al (2015) Elucidating the role of site-SPECIFIC Nitration of α-Synuclein in the
pathogenesis of Parkinson’s disease via protein semisynthesis and mutagenesis. J Am Chem
Soc 137(15):5041–5052
112. Kleinknecht A et al (2016) C-terminal tyrosine residue modifications modulate the protective
phosphorylation of serine 129 of α-synuclein in a yeast model of Parkinson’s disease. PLoS
Genet 12(6):e1006098
113. Hodara R et al (2004) Functional consequences of α-synuclein tyrosine nitration: diminished
binding to lipid vesicles and increased fibril formation. J Biol Chem 279(46):47746–47753
114. Li X et al (2012) Iron increases liver injury through oxidative/nitrative stress in diabetic rats:
involvement of nitrotyrosination of glucokinase. Biochimie 94(12):2620–2627
115. Shavali S, Combs CK, Ebadi M (2006) Reactive macrophages increase oxidative stress and
α-synuclein nitration during death of dopaminergic neuronal cells in co-culture: relevance to
Parkinson’s disease. Neurochem Res 31(1):85–94
116. Reynolds AD et al (2008) Nitrated α-synuclein-activated microglial profiling for Parkinson’s
disease. J Neurochem 104(6):1504–1525
117. Wen L et al (2018) NLRP3 inflammasome activation in the thymus of MPTP-induced Parkin-
sonian mouse model. Toxicol Lett 288:1–8
118. Yu Z et al (2010) Nitrated α-synuclein induces the loss of dopaminergic neurons in the
substantia nigra of rats. PLoS ONE 5(4):e9956
119. Fujiwara H et al (2002) α-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell
Biol 4(2):160–164
120. Anderson JP et al (2006) Phosphorylation of Ser-129 is the dominant pathological modification
of α-synuclein in familial and sporadic Lewy body disease. J Biol Chem 281(40):29739–29752
121. Chen L, Feany MB (2005) α-synuclein phosphorylation controls neurotoxicity and inclusion
formation in a Drosophila model of Parkinson disease. Nat Neurosci 8(5):657–663
4 Iron Pathophysiology in Parkinson Diseases 63
122. Huang B et al (2018) Phosphorylated α-synuclein accumulations and Lewy body-like pathol-
ogy distributed in Parkinson’s disease-related brain areas of aged Rhesus monkeys treated
with MPTP. Neuroscience 379:302–315
123. Kuwahara T et al (2012) Phosphorylation of α-synuclein protein at Ser-129 reduces neuronal
dysfunction by lowering its membrane binding property in Caenorhabditis elegans. J Biol
Chem 287(10):7098–7109
124. Oueslati A et al (2013) Polo-like kinase 2 regulates selective autophagic α-synuclein clearance
and suppresses its toxicity in vivo. Proc Natl Acad Sci U S A 110(41):E3945–E3954
125. Sancenon V et al (2012) Suppression of α-synuclein toxicity and vesicle trafficking
defects by phosphorylation at S129 in yeast depends on genetic context. Hum Mol Genet
21(11):2432–2449
126. Tenreiro S, Eckermann K, Outeiro TF (2014) Protein phosphorylation in neurodegeneration:
friend or foe? Front Mol Neurosci 7:42
127. Takahashi M et al (2007) Oxidative stress-induced phosphorylation, degradation and aggre-
gation of α-synuclein are linked to upregulated CK2 and cathepsin D. Eur J Neurosci
26(4):863–874
128. Perfeito R et al (2014) Linking α-synuclein phosphorylation to reactive oxygen species for-
mation and mitochondrial dysfunction in SH-SY5Y cells. Mol Cell Neurosci 62:51–59
129. Lu Y et al (2011) Phosphorylation of α-Synuclein at Y125 and S129 alters its metal binding
properties: implications for understanding the role of α-Synuclein in the pathogenesis of
Parkinson’s Disease and related disorders. ACS Chem Neurosci 2(11):667–675
130. Cahill CM et al (2009) Amyloid precursor protein and alpha synuclein translation, impli-
cations for iron and inflammation in neurodegenerative diseases. Biochim Biophys Acta
1790(7):615–628
131. Sofroniew MV, Vinters HV (2010) Astrocytes: biology and pathology. Acta Neuropathol
119(1):7–35
132. Mirza B et al (2000) The absence of reactive astrocytosis is indicative of a unique inflammatory
process in Parkinson’s disease. Neuroscience 95(2):425–432
133. Heneka MT, Rodriguez JJ, Verkhratsky A (2010) Neuroglia in neurodegeneration. Brain Res
Rev 63(1–2):189–211
134. Neal M, Richardson JR (2017) Epigenetic regulation of astrocyte function in neuroinflamma-
tion and neurodegeneration. Biochim Biophys Acta 1864(2):432–443
135. Collins LM et al (2012) Contributions of central and systemic inflammation to the pathophys-
iology of Parkinson’s disease. Neuropharmacology 62(7):2154–2168
136. Long-Smith CM, Sullivan AM, Nolan YM (2009) The influence of microglia on the patho-
genesis of Parkinson’s disease. Prog Neurobiol 89(3):277–287
137. Phani S, Loike JD, Przedborski S (2012) Neurodegeneration and inflammation in Parkinson’s
disease. Parkinsonism Relat Disord 18(Suppl 1):S207–S209
138. Benarroch EE (2009) Brain iron homeostasis and neurodegenerative disease. Neurology
72(16):1436–1440
139. Rouault TA, Zhang DL, Jeong SY (2009) Brain iron homeostasis, the choroid plexus, and
localization of iron transport proteins. Metab Brain Dis 24(4):673–684
140. Healy S, McMahon JM, FitzGerald U (2017) Modelling iron mismanagement in neurode-
generative disease in vitro: paradigms, pitfalls, possibilities & practical considerations. Prog
Neurobiol 158:1–14
141. Zecca L et al (2004) Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci
5(11):863–873
142. Bartzokis G et al (2007) Brain ferritin iron may influence age- and gender-related risks of
neurodegeneration. Neurobiol Aging 28(3):414–423
143. Kress GJ, Dineley KE, Reynolds IJ (2002) The relationship between intracellular free
iron and cell injury in cultured neurons, astrocytes, and oligodendrocytes. J Neurosci
22(14):5848–5855
144. Oshiro S et al (2008) Microglia and astroglia prevent oxidative stress-induced neuronal cell
death: implications for aceruloplasminemia. Biochim Biophys Acta 1782(2):109–117
64 H. Jiang et al.
168. Lan J, Jiang DH (1997) Desferrioxamine and vitamin E protect against iron and MPTP-
induced neurodegeneration in mice. J Neural Transm (Vienna) 104(4–5):469–481
169. Febbraro F et al (2013) Chronic intranasal deferoxamine ameliorates motor defects and pathol-
ogy in the α-synuclein rAAV Parkinson’s model. Exp Neurol 247:45–58
170. Dexter DT et al (2011) Clinically available iron chelators induce neuroprotection in the 6-
OHDA model of Parkinson’s disease after peripheral administration. J Neural Transm (Vienna)
118(2):223–231
171. Shachar DB et al (2004) Neuroprotection by a novel brain permeable iron chelator, VK-28,
against 6-hydroxydopamine lession in rats. Neuropharmacology 46(2):254–263
172. Gal S et al (2010) Restoration of nigrostriatal dopamine neurons in post-MPTP treatment by
the novel multifunctional brain-permeable iron chelator-monoamine oxidase inhibitor drug,
M30. Neurotox Res 17(1):15–27
173. Mena NP et al (2015) The novel mitochondrial iron chelator 5-((methylamino)methyl)-8-
hydroxyquinoline protects against mitochondrial-induced oxidative damage and neuronal
death. Biochem Biophys Res Commun 463(4):787–792
174. Zhu W et al (2007) Prevention and restoration of lactacystin-induced nigrostriatal dopamine
neuron degeneration by novel brain-permeable iron chelators. FASEB J 21(14):3835–3844
175. Gal S et al (2006) M30, a novel multifunctional neuroprotective drug with potent iron chelating
and brain selective monoamine oxidase-ab inhibitory activity for Parkinson’s disease. J Neural
Transm Suppl 70:447–456
176. Gotsbacher MP et al (2017) Analogues of desferrioxamine B designed to attenuate iron-
mediated neurodegeneration: synthesis, characterisation and activity in the MPTP-mouse
model of Parkinson’s disease. Metallomics 9(7):852–864
177. Telfer TJ et al (2017) Adamantyl- and other polycyclic cage-based conjugates of desferriox-
amine B (DFOB) for treating iron-mediated toxicity in cell models of Parkinson’s disease.
Bioorg Med Chem Lett 27(8):1698–1704
178. Das B et al (2017) A Novel Iron(II) Preferring Dopamine Agonist Chelator as Potential
Symptomatic and Neuroprotective Therapeutic Agent for Parkinson’s Disease. ACS Chem
Neurosci 8(4):723–730
179. Wang N et al (2017) Iron chelation nanoparticles with delayed saturation as an effective
therapy for Parkinson disease. Biomacromol 18(2):461–474
180. Aguirre P et al (2017) Neuroprotective Effect of a new 7,8-dihydroxycoumarin-based
Fe(2+)/Cu(2+) chelator in cell and animal models of Parkinson’s disease. ACS Chem Neurosci
8(1):178–185
181. Abdelsayed S et al (2015) Piperazine derivatives as iron chelators: a potential application in
neurobiology. Biometals 28(6):1043–1061
182. Workman DG et al (2015) Protection from neurodegeneration in the 6-hydroxydopamine
(6-OHDA) model of Parkinson’s with novel 1-hydroxypyridin-2-one metal chelators. Metal-
lomics 7(5):867–876
183. Finkelstein DI et al (2017) The novel compound PBT434 prevents iron mediated neurodegen-
eration and α-synuclein toxicity in multiple models of Parkinson’s disease. Acta Neuropathol
Commun 5(1):53
184. Xu Q et al (2017) Epigallocatechin gallate has a neurorescue effect in a mouse model of
Parkinson disease. J Nutr 147(10):1926–1931
185. Zhao J et al (2017) Metal chelator EGCG attenuates Fe(III)-induced conformational transi-
tion of α-synuclein and protects AS-PC12 cells against Fe(III)-induced death. J Neurochem
143(1):136–146
186. Chan DK et al (1998) Genetic and environmental risk factors for Parkinson’s disease in a
Chinese population. J Neurol Neurosurg Psychiatry 65(5):781–784
187. Wang YQ et al (2015) Neuroprotective effects of ginkgetin against neuroinjury in Parkinson’s
disease model induced by MPTP via chelating iron. Free Radic Res 49(9):1069–1080
188. Ma ZG et al (2007) Myricetin reduces 6-hydroxydopamine-induced dopamine neuron degen-
eration in rats. NeuroReport 18(11):1181–1185
66 H. Jiang et al.
189. Haleagrahara N, Siew CJ, Ponnusamy K (2013) Effect of quercetin and desferrioxamine
on 6-hydroxydopamine (6-OHDA) induced neurotoxicity in striatum of rats. J Toxicol Sci
38(1):25–33
190. Du XX et al (2012) Curcumin protects nigral dopaminergic neurons by iron-chelation in the
6-hydroxydopamine rat model of Parkinson’s disease. Neurosci Bull 28(3):253–258
191. Guo C, Chen X, Xiong P (2014) Baicalin suppresses iron accumulation after substantia nigra
injury: relationship between iron concentration and transferrin expression. Neural Regen Res
9(6):630–636
192. Xiong P et al (2012) Baicalin and deferoxamine alleviate iron accumulation in different brain
regions of Parkinson’s disease rats. Neural Regen Res 7(27):2092–2098
193. Xu H et al (2010) Rg1 protects the MPP+ -treated MES23.5 cells via attenuating DMT1
up-regulation and cellular iron uptake. Neuropharmacology 58(2):488–494
194. Gonzalez-Burgos E, Fernandez-Moriano C, Gomez-Serranillos MP (2015) Potential neuro-
protective activity of Ginseng in Parkinson’s disease: a review. J Neuroimmune Pharmacol
10(1):14–29
Chapter 5
Iron Pathophysiology in Alzheimer’s
Diseases
Tao Wang, Shuang-Feng Xu, Yong-Gang Fan, Lin-Bo Li and Chuang Guo
Tao Wang, Shuang-Feng Xu, Yong-Gang Fan, Lin-Bo Li—These authors contributed equally to
this work
5.1 Introduction
memory disorders; that is, short-term events cannot be recalled. As the pathology of
AD progresses, the patient’s personality and behavior will change, severe cognitive
function declines, mental disorders appear, speaking and acting very difficult, and
losing self-care ability in the late stage, and following extremely high mortality from
multiple complications [9].
Pathologically, the typical pathological features of AD, the deposition of NFTs
formed by hyperphosphorylation of tau protein in neurons and extracellular amyloid-
β (Aβ) plaques, can be detected in early AD [167]. NFTs and Aβ are also detected
on PET scanning in healthy human brains, but normal organisms metabolize them or
allow them to dissolve in cell fluids to perform normal functions [167]. In the brain
of AD patients, these proteins are excessively present, and the clearance is blocked,
which affects cell survival along with synaptic and neuronal losses particularly in
cortex and hippocampus [9, 90] and hence atrophy of brain tissue. Hippocampus is
associated with memory function; hippocampal atrophy is regarded as an important
indicator of memory impairment. When performing neurological examination and
cranial CT or magnetic resonance imaging (MRI) in early AD, most of them have
positive findings [167]. As the pathology of AD progresses, the plaque and tangles
formed by the middle of the brain in the memory and thinking area of the AD patients
are more, resulting in impaired memory and thinking ability of the patient. At this
stage, plaque and tangles spread to the sensory area responsible for speaking and
understanding of the language and for the body to be in contact with the surrounding
objects. Its pathological manifestations are atrophy of the cerebral cortex, flatten-
ing of the brain, widening of the sulcus, enlargement of the ventricles, and weight
reduction of brain. The atrophy of the temporal lobe, parietal lobe, prefrontal cortex,
and hippocampus was the most obvious, and it was more significant than the early
patients. In the advanced stage of AD, most of the patient’s cerebral cortex has been
severely damaged, neurons have died, and the brain has severely shrunk [221, 282].
in the loss of function of several enzymes that require iron as a cofactor, forming
toxic oxidizing species, and increasing β-amyloid production [226]. Several com-
mon genetic polymorphisms that cause elevated levels of iron and disorders in the
body are associated with AD [63, 198], but the pathology is unclear.
It is widely believed that aging is a major risk factor for neurodegenerative dis-
eases. Iron will accumulate in several brain regions of healthy elderly people and will
exhibit a large degree of accumulation in the brains of patients with neurodegenera-
tive diseases [53, 55, 56, 119, 295]. In vitro, increasing iron concentration accelerates
Aβ plaque and pTau tangles and increases its toxicity [240]. Autopsy results showed
that total iron deposition was positively correlated with age, and altered iron levels
were observed in specific brain areas of AD patients (the hippocampus) compared
with the age-matched control group [232, 308]. Iron elevation in the AD brain was
first reported in 1953 [108], and its association with Aβ and NFTs or with ferritin in
peripheral glial cells was confirmed in some studies [55, 56, 150, 176, 295]. Clin-
ical studies have also shown that the disorder of iron metabolism is the key to the
pathology of AD. It is based on the observation that the patient’s content in the
cortex, subcortex, and white matter is significantly increased [233]. MRI analysis
showed that iron levels in the hippocampus increased in the early stage of AD and
were negatively correlated with memory test performance [68]. Moreover, the data
of MRI and histological analysis suggest that increased iron load in the brain is also
associated with Aβ plaque formation, which locally binds to the Aβ core and halo
regions and is entangled with hyperphosphorylated tau (pTau) in humans and animals
[107, 182, 260]. At the cellular level, oligodendrocytes require iron as a cofactor for
enzymes involved in myelin production and maintenance and have the highest iron
content of all cell types in CNS, which increases with age and even further in AD
[21]. Compared to other cells, oligodendrocytes contain low levels of anti-oxidants
[273], which renders them vulnerable to oxidative stress that can accompany a rapid
increase in intracellular iron [199]. Thus, the observation that neuroimaging-defined
white matter abnormalities are characteristic of AD is relatively new, which may be
radiological manifestations in preclinical AD [85, 199]. In addition, the successful
application of iron chelators in patients with early AD and several AD animal models
[62, 78, 115, 114, 116, 142, 237, 309] provided strong evidence that iron-overloading
in specific brain areas with age is associated with the pathophysiological progres-
sion of AD. Of note, an important consideration in studying iron levels in AD is
to emphasize that the increase in total iron content in the brain is not necessary for
brain oxidative stress alone, but that the body may favor oxidation of Aβ and NFTs
by inducing an iron-rich environment, causing agitation and cell death, while other
brain regions may suffer from impaired nerve function due to iron deficiency [24].
Iron is a necessary element demanded for virtually all living organisms and compli-
cates in numerous biological reactions such as electron transfer oxygen transport,
oxidative metabolism, and gene regulation. In recent years, numerous studies have
5 Iron Pathophysiology in Alzheimer’s Diseases 71
revealed that abnormally elevated brain iron is the cause of certain neurodegen-
erative diseases [160]. Nonetheless, why iron accumulates in these disorders and
other important questions regarding their pathogenesis remain unanswered [188].
The abnormally increased iron ions in the brain of AD patients can interact with
H2 O2 to produce highly active hydroxyl radicals in the body. Hydroxyl radicals act
on the proteins, nucleic acids, and cell membrane containing a large amount of unsat-
urated fatty acids and then cause cell damage, called oxidative stress damage [161].
Mitochondria are an energy processing plant in cells. It is also an important site
for generating free radicals. Excessive iron destroys the mitochondrial respiratory
chain, the destruction of any part of the respiratory chain causes the formation of
free radicals, and then the biological macromolecules are damaged, eventually the
cells death [8, 131]. Accumulated iron can also damage mitochondrial membrane
potential, activate caspase-3-dependent apoptotic pathway leading to cells apoptosis,
and ultimately lead to the occurrence of AD [164].
In AD, Fe accumulation has been observed in and around the Aβ, SPs and NFTs
[260]. Cerebral iron deposition may be related to the metal-binding sites of Aβ
and tau proteins and may be related to abnormal expression of various brain iron
metabolism-related proteins in the brain of AD patients.
Interactions between iron and Aβ or APP
APP is expressed in most cell types, both brain-derived and other tissues, including
blood cells [238]. In the amyloidogenic pathway, APP is a transmembrane met-
alloprotein [194] that is cleaved by β-secretase enzyme and γ-secretase enzyme
to generate the neurotoxic 40–42-amino-acid prefibrillar amyloid [286]. During the
non-amyloidogenic path of APP metabolism, α-secretase enzyme is the zinc metallo-
protease that cleaves APP in the Aβ-peptide domain to release the 90 kDa ectodomain
of APP. According to reports, we know that α-secretase activation produces a 90 kDa
APP(s) and APP mRNA translation upregulation after interleukin (IL)-1-mediated
stress response, ultimately resulting in secretion of the APP, is increased [19]. Some
study findings showed that APP provides protection against catalyzed oxidative
stress, for example, the lesions during the pathogenesis of AD or lesion and reper-
fusion after stroke [91].
Aggregation of Aβ in brain is the main pathological characteristic of AD, which
is known to be triggered by metals such as iron. Aβ can directly bind iron through
its His6, His13, His14, due to the universality of the binding of ferrous iron to
Aβ, and the interaction between Aβ and iron is believed to be beneficial to smaller
microenvironments such as the brain [32, 298]. Recently, a study uncovering the
relationships between iron biochemistry and AD pathology in APP/PS1 mouse cortex
revealed that iron is an integral part of the SP and also provided the evidence that
Aβ-induced reduction of iron can be occurred in vivo [269]. The iron-aggregated Aβ
72 T. Wang et al.
Iron plays an important role in the brain. On the one hand, it maintains the brain’s
extremely high respiratory activity for the benefit of myelination and the produc-
tion of various neurotransmitters [37]. On the other hand, due to the high oxygen
metabolism rate of brain tissue and metal ions such as iron, and the relative lack
of antioxidant protection mechanism, it is easy to cause a loss of balance between
oxidative damage and antioxidant defense of brain tissue [49]. Oxidative stress is
a common pathological feature inside the development and pathogenesis of sev-
eral neurodegenerative diseases, which includes AD [161, 291]. Empirical evidence
shows that oxidative stress occurs prior to the advent of SPs and NFTs, thus leading to
the signs and symptoms of dementia [291]. Clinical data further prove an association
between oxidative stress and AD-like cognitive dysfunction [140]. Oxidative stress
contributes to the production of Aβ and hence aggravates its neurotoxicity [193].
In recent years, lots of studies have confirmed that substantially all intracellular
macromolecules, such as lipid, protein and nucleic acid, existing in oxidized form
in the brain tissue of AD patients, and the level of oxidized form are significantly
increased.
Lipid peroxidation
It has been excellently revealed that lipid peroxidation comprises three distinct mech-
anisms, free radical-mediated oxidation, free radical-independent, nonenzymatic oxi-
dation, and enzymatic oxidation [205, 250, 256]. Lipid peroxidation has been shown
to induce disturbance to membrane organization and functional disorder of proteins
and DNA. However, lipid peroxidation products also act as redox signaling mediators
[311].
The aggregation of Aβ is recommended to play a central role in the pathogenesis
of AD which is the most common cause of dementia, Aβ, and small oligomers can
insert into the lipid bilayer and generate hydrogen peroxide [135]. In the presence
of redox-active transition metal ions, consisting of iron and copper, hydrogen per-
oxide undergoes a Fenton reaction resulting in the formation of hydroxyl radicals.
The reaction then develops lipid peroxidation and causes oxidative impairment to
proteins and biomolecules [40]. On the other hand, using an animal model of AD,
it has been shown that oxidative stress and lipid peroxidation can induce Aβ accu-
mulation during the progression of AD [223]. The growing body of evidence for the
involvement of lipid peroxidation and general oxidative stress as a driving force of
AD has generated much interest in using antioxidants as therapeutics [94]. Since the
antioxidant capacity of a cell is dependent on a cocktail of multiple antioxidants, it
is possible that supplementation with one or two antioxidants is not sufficient for
changing the course of disease in humans. Additionally, since lipid peroxidation can
be initiated at very early stages of the disease, antioxidant therapy may be more
effective in patients with early stages of AD, rather than late-stage patients [218].
74 T. Wang et al.
In the foregoing text of the present article, we have already mentioned that the brain
is susceptible to oxidative stress and ROS caused by the presence of iron. Homeosta-
sis of iron is tightly regulated under normal physiological conditions as excessive
amounts of iron are known to cause cellular susceptibility to oxidative stress excess,
and then injury DNA and/or RNA via ROS mechanism [128]. The accumulation of
DNA and RNA oxidative damage is observed in cortical and hippocampal neurons
from AD brains at early stages of pathology [283]. An increase in oxidative DNA and
RNA damage occurs in a subset of cortical and hippocampal neurons at the onset of
AD [33]. Non-repaired nucleic acid damage is highly deleterious in neurons, as it can
trigger transcriptional or translational dysregulation and chromosomal instability, as
observed in AD [58]. However, it is poorly comprehended about the mechanisms
underlying the alteration of nucleic acid integrity in neurons all through the early
tiers of AD pathogenesis [128]. In unique, the capability role of tau pathology in
nucleic acid integrity has not been explored.
Several groups have documented the accumulation of oxidative DNA damage
during aging in the brain [121, 127]. Oxidative damage to DNA may be one of the
maximum crucial elements in neuron degeneration of AD due to the critical role of
DNA in cell function. Lu et al. [171] located that oxidative DNA damage preferen-
tially accumulates in the promoter regions of several genes concerned in synaptic
plasticity, vesicular transport, and mitochondrial function. It has been mentioned that
differential accumulation of oxidative DNA damage in the brain regions indicates
that certain regions of the brain are more susceptible to oxidative DNA modifications
than other regions [42, 101]. Without efficient repair ability in the brain in AD, muta-
tions in nDNA and mtDNA may result in neuron death through defects in oxidative
phosphorylation [93, 170]. Furthermore, higher concentrations of oxidized pyridines
and purines were detected in lymphocytes and leukocytes of AD patients compared
with controls [185, 190], and increased levels of 8-oxoG were observed in DNA from
ventricular CSF of AD patients [170]. Recent findings indicate increased oxidative
damage in leukocytes [190] and brain tissue [288] of subjects with MCI, indicating
that DNA oxidation may constitute an early occasion in the AD progression, prior to
cytopathological changes, and might make contributions to the pathogenesis of the
disease [184].
Abundant evidence considerably supports the early involvement of RNA oxidation
in the pathological of neurodegeneration, particularly in AD. RNA oxidation was
observed in postmortem brains of early AD cases [208], a presymptomatic case
with familial AD mutation [207]. Oxidized RNAs may demote faster than other
types of oxidatively modified macromolecules, while protein carbonyl compounds,
lipid peroxidation products, and glycoxidation products are stable at the point of
production [210]. Researches proved the observations contrast remarkably with the
pattern of the RNA oxidation, a steady-state marker, which is prominent in neurons
without pathology [208, 209].
As for RNA oxidization, a study has shown that the Fenton reaction oxidized
and cleaved rRNA in cultured primary neurons and astrocytes [69]. It is known that
5 Iron Pathophysiology in Alzheimer’s Diseases 75
redox-active iron has been shown to also bind with RNA at the metal-binding site
[25] and that redox-active iron bound to ribosomes oxidizes rRNA in AD [130]. Of
course, mRNA oxidation is not a negligible event and may play an important role in
the pathogenesis of AD [255].
Mitochondrial dysfunction
Mitochondrial dysfunction has long been associated with several neurodegenerative
diseases including AD [278]. Mitochondria are essential for regulating the equilib-
rium of iron throughout the cell [219]. Mitochondrial function is critical for normal
cellular processes. It is particularly notable that many neurodegenerative diseases are
directly related to iron metabolism disorders in mitochondria [278, 293]. For exam-
ple, an abnormal increase in iron in brain tissue of patients with AD and PD is likely
to be one of the important causes of neuronal death [20]. Mitochondrial damage and
oxidative stress have been greatly implicated in the progression of aging, along with
the pathogenesis of age-related neurodegenerative diseases, such as AD, PD, and
ALS [79]. Several mitochondria-targeted strategies, such as promoting antioxidant
bioavailability through novel delivery systems, figuring out unique mitochondrial
proteins as precise drug targets, studying signaling pathways for mitochondrial bio-
genesis and kinetics, and identifying effective natural products may be effective
resistant to neurodegenerative diseases associated with mitochondrial dysfunction
[268].
In fact, the early stage of AD is identified via morphologically and functionally
impaired mitochondria, which are prone to over-form ROS, resulting in reduced
brain energy due to ATP reduction [102]. Once formed, Aβ corresponds with the
mitochondria and causes further mitochondrial defect. Aβ inhibits complex IV and
binds Aβ-bound alcohol dehydrogenase (ABAD). ABAD produces ROS. It has been
reported that mitochondria contain an active γ-secretase complex that is involved in
processing of APP to form Aβ, and the non-soluble APP can affect outer mitochon-
drial membrane and interfere with protein shuttle import [102]. Impaired crosstalk
between endoplasmic reticulum and mitochondria also seems to play an important
role in AD pathogenesis. In addition, mitochondrial dysfunction and apoptosis are
associated with Aβ-mediated toxicity [251]. In brief, mitochondria function as both
the source and target of toxic ROS because mitochondrial dysfunction and oxida-
tive stress are important in age-related neurodegenerative diseases, particularly AD
[281]. However, whether mitochondrial iron accumulation is a founding event or a
consequence of upstream events in a particular disease is a matter of debate [187].
5.4.3 Neuroinflammation
NMDARs, a cation channel, adjust the entry of Na+ and Ca2+ ions when are activated
by the co-agonists glutamate (or NMDA) and glycine or D-serine. Appropriate acti-
vation of NMDARs plays a critical role in physiological functions such as excitatory
neurotransmission, synaptic plasticity [45]. Activated NMDARs cause speedy open-
ing of Ca2+ channels on the cell membrane, followed by an increase in free Ca2+ ions
in the cytoplasm with the aid of a CNS signaling cascade [215]. However, excitotoxic-
ity induced by excessive activation of NMDARs contributes to pathological changes
in the CNS [106]. Hippocampal neurons require iron to generate calcium signals
after NMDARs’ stimulation [196]. Studies of NMDARs involvement in the NTBI
influx pathways implied that activation of NMDARs drastically promotes Fe2+ entry
into cells in primary neurons [48, 216]. Moreover, iron has been shown to block
the influx of Ca2+ in the NMDARs channel, suggesting that Fe2+ competes with
Ca2+ for NMDARs in cultured neurons [197]. In addition, NMDAR’s activation has
been mentioned to promote iron accumulation by accelerating DMT1-mediated iron
influx [48], amplifying iron releasing from lysosome and regulating DMT1 expres-
sion [296]. Interestingly, it is the increased expression of DMT1+IRE mRNA rather
than DMT1-IRE mRNA after treatment of NMDA in primary hippocampal neurons
[118]. In fact, iron deficiency can inhibit cognitive ability [43, 81, 196]; thus, it is of
potential value to ensure an adequate iron supply through the pathway of iron entry
from DMT1. This means that NMDAR’s activation may contribute to the formation
of hippocampal spatial memory by stimulating the expression of DMT1+IRE.
Of note, a role for iron in NMDAR-mediated synaptic plasticity has begun to
remind. A previous study suggest that ROS generated by iron are required for
NMDA-induced and RyR-mediated ERK1/2 phosphorylation/nuclear translocation
(a requisite step of sustained synaptic plasticity) in primary hippocampal neurons,
and electrophysiological studies also found that iron addition resulted in a signifi-
cant increase of the long-term potentiation (LTP), but the increase can be prevented
by iron chelation [196]. Recent studies reported that NMDARs were substantially
improved within the prefrontal cortex and hippocampus of iron-loaded rats, sug-
gesting that altered iron status by iron supplementation in the brain regulates gluta-
matergic neurotransmission pathway [122]. In the hippocampus, studies found that
intracellular iron signaling could modulate neuronal excitability [296], suggesting
that iron can make in a feedback method to modulate NMDA function to maintain
neuronal excitability under normal condition. Further study showed that iron-induced
ROS generation is required for Ca2+ release for long-term potentiation in primary hip-
pocampal neurons [196]. However, iron overload might promote NMDARs mediated
excitotoxicity possibly by enhancing Ca2+ release and calcium-induced cell damage.
Both iron dyshomeostasis and NMDAR-mediated neurotoxicity have been shown to
have an important role in neurological diseases such as AD [301].
78 T. Wang et al.
Synaptic loss and neuronal death are one of the major pathological hallmarks of
AD, and the endurance of neuronal loss is correlated with the progression of the
disease. Synapses are formed by the junction between two neurons, which allows
neuronal cells to transmit signals to another cell. This channel is usually damaged
or lost in many neurodegenerative diseases, such as AD [143, 257]. Accumulat-
ing evidence suggests that dysfunction and loss of synaptic connections may be
an important early event underlying AD progression, more than other pathological
changes [12, 57, 262]. Damage to synapses by Aβ oligomers includes changes in
synaptic numbers, synaptic transmission, and changes in synaptic plasticity [80, 224,
262]. Comparably, evidence also shows that pathological changes in tau are toxic for
synapses [222]. The molecular mechanisms that lead to synapse degeneration and
neuronal loss downstream of Aβ and tau are less well established, but different path-
ways are implicated such as metal dyshomeostasis, oxidative stress, mitochondrial
dysfunction, and neuroinflammation [64]. Some results suggested that iron impacts
mitochondrial dynamics, possibly trigging synaptic loss and apoptotic, and suggests
that iron chelators have to take into consideration as a capability molecule with
memory restoring and neuroprotective properties, observed in the remedy of cogni-
tive deficits in neurodegenerative disorders [259, 117]. Reduced synaptophysin may
be associated with iron-induced cognitive deficits observed in AD model, as these
researchers have previously reported that iron-induced hippocampal synaptophysin
levels continue to decline in rats [259, 155]. Recently, some data on iron deficit that
may play a role in synaptic processes, as well as preceding studies on the effects of
iron deficiency on cognitive processes and synaptic function, can generate valuable
information to prevent against the effects of iron deficiency and iron overload [195].
Thus, insightful synapse degeneration in AD is characterized by the worsening of
cognitive function, synapse loss, and neuronal cell death.
Morphologically, advanced AD manifests postmortem as a significant shrinkage
in cortical volume, especially throughout the cortex and hippocampus. This is caused
by massive neuronal cell death, whose etiology is not clear [88]. Notably, the type of
cell death that occurs in affected areas of the AD brain is still contentious, despite the
demonstration that DNA fragmentation and upregulation of pro-apoptotic proteins
has been frequently observed [227]. As such, it remains unclear to what extent apop-
tosis or emerging types of regulated necrosis are responsible for bulk neuronal loss
in AD [16, 227]. We now know that abnormal elevation of iron may be the initiat-
ing factor of neuronal death in the brain of patients with neurodegenerative diseases
[225]. Iron-induced oxidative stress has been shown to initiate several apoptotic sig-
naling pathways in neurons [243] and cause oxidative damage to key proteins. There
is extensive evidence that oxidative damage to proteins and lipids by iron can cause
synaptic dysfunction and neuronal cell death [178], both of which are critical features
of AD.
Ferroptosis is the fourth cell death pattern proposed by scholars based on the
unique pathological state caused by intracellular iron accumulation, which is differ-
5 Iron Pathophysiology in Alzheimer’s Diseases 79
ent from necrosis, apoptosis, and autophagy [73]. Ferroptosis signs a newly located
form of cell death that is not associated with the caspase pathway and is involved in
iron disorders, lipid peroxidation, and inflammation as predominant markers [72].
The disorder in cellular redox homeostasis leads to accumulation of lipid ROS and
hence lipid peroxidation [1, 97]. Morphological changes affecting the outcomes
involve mitochondrial volume reduction, reduced density of mitochondrial mem-
branes, chromatin condensation, eventually rupture of mitochondrial outer mem-
brane, and plasma membrane [73, 89]. Several powerful ferroptosis inducers have
been identified [267] in which the small-molecule erastin effectively inhibits xCTcys-
tine back transporter to further induce ferroptosis, as cystine is vital for glutathione
synthesis. Glutathione peroxidase 4 (GPX4) [305] utilizes reduced glutathione to
detoxify phospholipid hydroperoxides and hydrogen peroxides [279]. It should be
noted that GPX4 knockout indicates iron imbalance, lipid peroxidation, and inflam-
mation in mice, at the same time shows early signs of AD phenotype, including
behavior defect and hippocampal neurodegenerative disease [120, 253]. Intriguingly,
small-molecule inhibitors of ferroptosis can mitigate neurodegeneration and inflam-
mation observed in GPX4 knockout mice, suggesting that ferroptosis is a potential
neurodegenerative mechanism affecting neurons important for spatial function [120].
Over the past several years, neuroimaging studies have implicated micro- and
macrostructural abnormalities in white matter in the risk and progression of AD
[34, 35, 157]. Analysis of postmortem brain tissue of AD patients found out a chem-
ical change in white matter compared to non-demented patients: The amounts of
general protein, myelin basic protein, myelin proteolipid protein, cyclic nucleotide
phosphohydrolase, and cholesterol are notably reduced, indicating myelin loss [239].
The abnormal white matter in AD is mainly manifested in changes in myelin and
oligodendrocytes [41, 199]. Myelin loss and the inability of the oligodendrocytes,
the cells responsible for the production and maintenance of myelin, to repair myelin
damage may be additional central features of AD [41]. Due to the essential role of
oligodendrocyte lineage in the process of myelination and remyelination, changes in
the variety of oligodendrocytes or their precursor cells and/or their dysfunction can
affect myelin integrity and thus may be related to AD pathogenesis [179].
In the brain, iron is a key substance involved in the process of myelin protein and
fatty acid synthesis in oligodendrocytes. Iron deposition in the brain of AD patients,
which in turn causes an increase in oxidative stress, may result in increased cell
damage. It is reported that myelinating oligodendrocytes contains a lower amount of
antioxidant than other glial cells [141]. Therefore, high iron content and low antiox-
idant content make oligodendrocytes one of the most susceptible to oxidative stress
in the CNS [276]. Rather, iron deficiency during infancy can lead to neurocognitive
developmental delay, impaired learning and memory and in some cases can lead to
mental illness, in part due to impaired myelination [228]. Due to normal aging, loss
80 T. Wang et al.
of white matter increases free iron, which is toxic to the brain. MRI analysis showed
that white matter damage in AD patients preceded gray matter damage compared
with age-matched controls [3].
Ferritin
Mitochondrial ferritin
of amino acids. The inner cavity of FtMt and H-ferritin shares a highly conserved
sequence and a fully overlapped crystallographic structure [153], suggesting that
these two proteins have similar functions. Unlike ferritin, the expression of FtMt
is tissue-specific, and it is predominantly expressed in the testis and brain [244].
The major bio-function of FtMt is also sequestering excess free iron. Although the
ferroxidase activity of FtMt is not as strong as H-ferritin, the iron sequestering ability
is high. Overexpression of FtMt in Hela cells resulted in reduced expression of
cytosolic ferritin and cytosolic iron deficiency [61]. Elevated expression of FtMt
was observed in the brains of AD patients and was associated with anti-oxidative
activity [304]. Furthermore, FtMt deletion exacerbated Aβ-induced neurotoxicity
[292], whereas FtMt overexpression attenuated Aβ-induced neurotoxicity [300].
Lactoferrin
In 1939, the lactoferrin (Lf) was firstly identified in bovine milk and subsequently
isolated in 1960 from both human and bovine milk. Human Lf is a glycoprotein
which consists of 691 amino acids, which is constitutively expressed and secreted
by glandular epithelial cells and by neutrophils following induction. The Lf pos-
sesses multi-functional bioactivities, such as anti-microbe, anti-inflammation, and
immunomodulation; meanwhile, it is an iron-binding glycoprotein, which is impor-
tant for the modulation of iron homeostasis. Like Tf, Lf has been shown to reversibly
bind iron as Fe3+ with high affinity (K D ∼ 10−22 M), as well as retain Fe3+ until pH
values less than 3.0. The Fe3+ -binding activity of Lf is dependent on the concomitant
and synergistic binding of carbonate, CO3 2− [5]. Spectroscopic studies and the 3D
structure suggest that Lf binds the CO3 2− firstly, thus neutralizing the positive charge
of the arginine residue (Arg-121 in the N-lobe and Arg-465 in the C-lobe) [18]. The
participation of the CO3 2− in the iron binding to Lf is important for iron reversible
binding since the protonation of the CO3 2− is likely a first step to break up the iron
site at low pH values [2]. Lf has two main conformational states, the open metal-free
(apo-lactoferrin, apo-Lf) state and the closed metal-bound (holo-lactoferrin, holo-Lf)
state. Metal binding and release are determined by the large-scale conformational
changes of Lf, by which the domains open to release the irons or close over the
bound irons [99]. The metal-free apo-Lf form is flexible and more prone to thermal
denaturation and digestion by proteases. However, the metal-bound holo-Lf is con-
formationally rigid and very stable, and the bound irons can only be removed by
the very low pH or some iron-chelating reagents. Of note, the iron-binding activ-
ity of Lf is not specific, and other trivalent metal ions such as Ga3+ and Al3+ can
also bind to Lf, although the affinity is lesser than Fe3+ [17]. The functions of Lf
in AD pathology remain largely unknown. A recent study reported that intranasal
human Lf effectively inhibits the cognitive decline of APP/PS1 mice via activation
of ADAM10 expression [116]. Meanwhile, salivary Lf was identified as a diagnostic
biomarker for mild cognitive impairment and AD due to it is more accurate than the
cerebrospinal fluid total tau and Aβ42 [44].
82 T. Wang et al.
Hepcidin
Ferroportin
FPN, encoded by the SLC40A1 gene, a 571 amino acid transmembrane protein,
plays a critical role in iron metabolism. Although cells possess several mechanisms
to uptake iron, FPN is the only known cellular iron exporter [125]. Iron efflux is in the
form of ferrous iron; therefore, the activity of ferroxidase is necessary for FPN. The
ferroxidase converts the divalent iron into ferric iron, which is mainly accomplished
by the positive membrane iron transport accessory proteins or ceruloplasmin. Fpn
controls iron entry into the plasma through duodenal enterocytes (1–2 mg/day) or
macrophages that recycle near 20–30 mg/day of iron from senescent erythrocytes
[74]. Recently, FPN has been recognized as the receptor for hepcidin, a master reg-
ulator of iron homeostasis [235]. Upon binding to hepcidin, FPN is internalized and
subsequently submitted to lysosomal degradation [201], which results in a reduc-
tion of plasma iron concentration and an increase of intracellular ferritin. The FPN
expression in AD brain is reduced with uncertain reasons [229]. Considering that APP
stabilizes the cell-surface FPN expression [75] and substantial APP variants were
observed in AD patients, the decreased FPN expression may be highly associated
with APP mutations.
5 Iron Pathophysiology in Alzheimer’s Diseases 83
iron metabolism. However, the in vitro data all suggested that MTf does not play a
vital role in Fe transport and metabolism [149, 234]. In addition, the MTf knockout
mice showed normal fertility and development, with no morphological or histolog-
ical abnormalities [77, 254]. Taken together, although the high-affinity Fe-binding
site of MTf is structurally and functionally similar to that in Tf, MTf is not necessary
in Fe transport and metabolism. It has been reported that MTf is expressed in reactive
microglia and the secreted MTf is increased in sera in AD patients [148]. To date,
the function of MTf in AD is largely unknown, which need to be further elucidated.
HFE
HFE is a protein that comprised of 343 amino acids, which interacts with β2 M to
form a heterodimer expressed at the cell surface [83]. HFE contains a signal peptide,
an extracellular transferrin receptor-binding region (α1 and α2), an immunoglobulin-
like domain (α3), a transmembrane region, and a short cytoplasmic tail [83]. Gly-
cosylation of HFE is important for its normal intracellular trafficking and functions.
HFE can be glycosylated at residues N110, N130, and N234 during transport to the
cell membrane [26]. HFE plays an important part in the regulation of iron homeosta-
sis. In iron-loaded HFE knockout mice, liver hepcidin was significantly decreased
[4]. Furthermore, iron overload was rescued in HFE knockout mice with constitutive
overexpression of hepcidin [204]. These data suggested that HFE acts as an upstream
regulator of hepcidin expression. HFE can interact with TFR1 and TFR2 in the cell
membrane, and any changes in the expression of either HFE or TFR2 can cause
the downregulation of hepcidin [111, 146]. The binding affinity of HFE for TFR
is similar to the binding affinity of iron-loaded TF, and the HFE can competitively
bind to the iron-loaded TF-binding site of TFR, thereby inhibiting the iron uptake
[84]. The associations between HFE polymorphisms and AD were investigated in
some studies. The HFE H63D mutation is associated with prolonged endoplasmic
reticulum stress and increased the risk of AD [165, 175]. The clear functions of HFE
in AD pathology remain unexplored, possibly involved in cholesterol modulation
and iron regulation [6].
HO-1
HO-1 is one of the three isoforms of heme oxygenase (HO) family which act as
key rate-limiting enzyme in the degradation of iron-containing heme [173]. It is
widely distributed in many mammalian tissue cells and is also recognized as heat
shock protein 32 (Hsp32). The level of HO-1 is low at basic conditions and can
be highly increased in the response of numerous pro-oxidant agents and stimuli
including heat shock, heavy metals, ultraviolet rays and cytokines. After induction,
HO-1 plays a pleiotropic, protective role and shows antioxidant, anti-apoptotic, and
anti-inflammatory effects in various conditions [168]. The cytoprotective effects of
HO-1 depend on multiple cellular processes including the generation of bilirubin,
an antioxidant molecule, from the degradation of heme; the induction of ferritin,
a strong chelator of free molecular iron; and the liberation of CO, responsible for
HO-1 major anti-inflammatory and anti-apoptotic effects [274]. Ninety percent of
5 Iron Pathophysiology in Alzheimer’s Diseases 85
the body’s iron is derived from the iron of aging red blood cells, which is the iron
produced by the decomposition of hemoglobin. Therefore, the role of HO-1 in iron
metabolism is crucial. Studies showed that sustained HO-1 activity decreased the
expression of transferrin receptor-1 and increased the proteasomal degradation of
ferritin [162]. Because of the distinct transcriptional regulation hemoglobin have
from iron, the change of HO-1 also affect FPN1 and hepcidin [177]. Besides, HO-1
is also related to neurodegeneration diseases [247]. The expression levels of HO-1 in
reactive astrocytes are markedly increased in the hippocampus and cerebral cortex of
patients with AD relative to age-matched, non-demented controls [249]. In AD mice,
long-term overexpression of HO-1 causes phosphorylation of tau protein, which
accumulates in the brain and forms neuronal fiber tangles, aggravating the condition
[137]. To review most of the literature, the glial HO-1 response may serve as a robust
transducer of noxious stimuli, an important driver of relevant neuropathology and a
potentially disease-modifying therapeutic target [248].
Lipocalin-2
Lipocalins are small secreted proteins that play roles in many processes including
retinol transport and prostaglandin synthesis. The lipocalins are capable of binding
low-molecular-weight ligands due to a common secondary and tertiary structure
corresponds to a single, eight-stranded antiparallel β-barrel around a central pocket
[46]. Lipocalin-2 (LCN2), also known as oncogene 24p3 or neutrophil gelatinase-
associated lipocalin (NGAL), is a 25 kDa soluble and secretory protein and reportedly
functions as a mediator of inflammation and iron ion transport [191]. Studies showed
that LCN2 mediates iron ion transport by association with a siderophore. LCN2
saturated with iron (holo-form) can increase intracellular iron levels by moving and
releasing iron into the cytoplasm. Iron-free form of LCN2 (apo-form) can deplete
intracellular iron and transport it to the extracellular space via its receptor NGAL-R
[66]. It can increase or decrease intracellular iron content based on the body iron store,
making it a malleable factor in the maintenance of iron homoeostasis [47]. Of note,
LCN2 protein levels in blood increase with age and mild cognitive impairment [51]
and are increased in human postmortem brain tissues of AD [65, 200]. In the context
of AD pathology, in vitro studies demonstrated that astrocytes generate high levels
of LCN2 in response to Aβ and that LCN2 sensitizes brain cells more vulnerable to
Aβ-induced cell death [189, 200]. However, more insight into such effects of LCN2
using LCN2 KO mice is required to understand its importance in AD and its potential
as a therapeutic target [65].
Furin
showed that hepcidin is produced in the liver and processed into the mature active
peptide by furin. The hepcidin gene encodes a precursor prepropeptide of 84 amino
acids. Furin recognizes the consensus sequence and participates the two sequential
cleavages to produce the mature peptide of 25 amino acids [280]. Few studies point
out that furin plays an important role in the activation of both α- and β-secretase. Both
ADAM10 and ADAM17 (the α-secretase) contain a consensus furin motif which is
essential for their activation. The β-secretase, known as BACE, also requires prote-
olytic removal of its proregion by furin at a minimal furin site [271].
The role of iron in the pathology of AD has only received attention in the past few
decades, in part because of inconsistencies in the literature regarding whether iron
increase is a major or minor event in AD brain [252]. Moreover, iron deficiency
during infancy can lead to neurocognitive developmental delay, impaired learning,
and memory and in some cases can lead to mental illness, in part due to impaired
myelination [228]. However, an early inductively coupled plasma mass spectrometry
(ICPMS) study showed that even in the normal brain region, there is a regionally
specific change in metal content [230]. Thus, it is important to distinguish between
large organizations and specific areas of research for the iron status in tissues of AD
patients relative to age-matched controls, including the blood, cerebrospinal fluid
(CSF), and brain tissue.
Using ICPMS technology, Gerhardsson et al. [98] detected CSF iron levels in 174
AD patients (122 females, 52 males) and 54 control subjects (36 females, 18 males).
There was no significant difference in CSF iron levels between the two groups, and
the same results were obtained by using the same method in different laboratories [30,
134]. Lavados et al. [154] used atomic absorption spectrometry (AAS) to detect iron
levels in the CSF of 7 AD patients and 12 control groups. There was no significant
difference in CSF iron levels between the two groups, consistent with the previous
study [192]. Interestingly, in the above studies, there was no significant difference,
but the CSF iron in the control group was slightly higher than the experimental
group. Taken together, the iron content in the CSF of AD patients did not change
significantly in its pathological process.
5 Iron Pathophysiology in Alzheimer’s Diseases 87
For iron levels in AD blood, the researchers used different experimental methods to
verify the iron content in serum, plasma and whole blood of healthy controls and
AD patients. Alimonti et al. [7] used inductively coupled plasma atomic emission
spectrometry (ICPAES) to detect serum iron levels in 53 AD patients (36 females, 17
males) and 124 control subjects (43 females, 81 males). They found that the serum
iron levels in the control group were higher than those in the AD group. Using ICPMS,
several research teams also detected that healthy controls have higher serum iron
levels than patients with AD [23, 27, 30]. However, Giambattistelli et al. [100] used a
turbidity immunoassay kit to determined serum iron levels in 107 AD patients and 52
healthy controls, and there was no significant difference in serum iron levels between
the two groups. Similar findings were detected by both Vitros 5.1 [275] and FerroZine
colorimetry [264, 265], respectively. Using AAS, the observation of several research
teams also confirmed that there was no difference in serum iron content between
normal people and AD patients [136, 192, 263], while Ozcankaya et al. (Ozcankaya
and Delibas [213]; Vural et al. [284]) detected that serum iron levels were significantly
increased in AD patients than in the control group. Different conclusions from various
studies may be due to different experimental methods, different data collection times,
individual sample differences, and different geographical distribution of samples.
Therefore, changes in iron levels in the blood may not be sufficient to diagnose AD.
Compared with blood and CSF, changes in iron content in the brain have reference
significance for the diagnosis of AD.
Iron has been extensively studied in different brain regions of AD patients, and the
results are different. Corrigan et al. [60] used ICPMS to detect hippocampal iron
levels in 12 AD patients (10 females, 2 males) and 12 control subjects (4 females,
8 males). The hippocampal iron levels in AD patients were significantly increased
compared to the control group. Gu et al. [113] used the same method to detect cortical
iron levels in 12 AD patients and 11 control groups and found that cortical iron levels
were significantly increased compared to the control group. House et al. [133] used
ICPAES to detect 5 AD patients (2 females, 3 males) and 5 control groups (2 females,
3 males). The iron levels of amygdala, corpus callosum, cingulate cortex, caudate
nucleus, entorhinal cortex, frontal cortex, globuspallidus, hippocampus, parietal cor-
tex, lateral temporal cortex, thalamus, subcortical frontal white matter, and temporal
white matter found that except for lateral temporal cortex, iron levels were signifi-
cantly increased compared to the control group, and the iron content in the other brain
regions did not change significantly. Srivastava et al. [266] used ICPMS to detect iron
content in the cerebellum and parietal cortex of 4 AD patients and 4 control groups
and found no significant difference between the two groups. Galazka-Friedman et al.
88 T. Wang et al.
[95] and Griffiths et al. [112] used AAS to detect iron content in hippocampus of
AD patients and healthy controls and found that the iron content in hippocampus of
AD patients increased slightly, but there was no significant difference. Andrási et al.
[11] used instrumental neutron activation analysis (INAA) to detect iron content in
the whole brain of 5 AD patients and 5 control groups. No significant difference
was found between the two groups, but Cornett et al. [59] using the same method
found that the iron content in multiple brain regions of AD patients was significantly
increased. From the results of autopsy, iron is unevenly distributed in the brain of
AD patients, and there are certain metabolic disorders, which may be involved in the
pathological process of AD. Although the results are different, in general, iron has
deposition in different brain regions of AD patients. This phenomenon may be one
of the causes of memory loss in AD patients. Therefore, the increase of iron content
in the brain may be one of the methods for the diagnosis of AD.
In fact, the diagnosis of AD is difficult because pathologic AD findings are not
sufficient for symptoms. To investigate AD pathological development and biomarkers
for early-stage AD identification, we learned that neuroimaging data do not directly
measure AD-related pathology but suggest neurodegeneration and achieved many
meaningful results; particularly, the noninvasive detectability of Aβ plaques in MRI
has so far been largely attributed to focal iron deposition accompanying the plaques
at preclinical stages of AD [105].
The issue of iron dysregulation has been the target of therapeutic attempts for the
treatment of AD. In this regard, iron chelation is a potential therapeutic strategy
in both oxidative stress and protein aggregation processes but remains a matter for
debate. For a metal chelator to effectively exert chelation, it must have the following
characteristics: (1) target iron-enriched areas without depleting transferrin-bound
iron from plasma; (2) remove chelated iron from iron accumulation sites or transfer
it to other biological proteins, such as circulating transferrin; (3) easy to penetrate
the cell membrane and blood-brain barrier (BBB); and (4) no side effects or minor
side effects on the body [28].
Several iron chelators including desferrioxamine (DFO) and clioquinol (CQ) have
been recently designed for their potential use in the treatment of AD [242]. DFO, a
hexadentate ligand with a higher binding affinity for Fe3+ than Fe2+ , is able to inhibit
the formation of β-pleated sheets of Aβ(1–42) and dissolve preformed β-pleated
sheets of plaque-like amyloid in vitro [132]. Despite a variety of promising results
in some clinical trials [62], experimental mouse models, the molecular size, and
hydrophilicity of DFO prevent it from freely crossing the BBB, when administered
orally [231]. Again, long-term administrated DFO leads many side effects, including
allergic reactions, anemia, liver and renal dysfunction, and neuronal hearing loss
[29]. Our recent studies demonstrated that intranasal DFO treatment inhibits iron-
induced amyloidogenic APP processing, tau hyperphosphorylation, and impaired
5 Iron Pathophysiology in Alzheimer’s Diseases 89
spatial learning and memory [115, 114], suggesting that intranasal administration of
DFO may be a safe and noninvasive therapeutic strategy for AD therapy. CQ, a small
lipophilic iron chelator that can cross the BBB, has been used in phase II clinical trials
for moderate AD cases [236]. However, CQ is not a iron selective and produces very
toxic effects. Surprisingly, recent research implicating the role of CQ in the inhibition
of cAMP efflux might represent promise for the treatment of neurodegenerative
diseases [217]. In addition, plant polyphenols were proposed for the treatment of
AD because of their antioxidant activities and strong iron-binding affinity [176],
but their capacity to be absorbed at the gastrointestinal tract and be transported
subsequently through the BBB is limited. Clearly, the development of an effective
non-toxic therapeutic agent for AD-like brain disorders remains a challenging task.
α-lipoic acid (LA), a small-molecule compound that can be naturally synthesized
in mammals, is mainly used for the treatment of diabetic polyneuropathy, and it
was recently found that 129 patients with AD who had been treated with LA for a
period of time had an effective remission of cognitive decline [82]. In fact, in vitro
studies have demonstrated the antioxidant properties of LA and a similar role as
metal chelating agents [212]. Studies showed that LA not only reduced iron uptake
rates but also decreases dynamic iron pools by increasing iron stores within the lens
epithelial cells [110]. In vivo, LA can enhance Aβ solubility the frontal cortex of APP
transgenic mice and human AD brain tissues, suggesting that LA, like other transition
metal chelators, reduces deposition of amyloid protein through Aβ re-dissolution
[86]. Most recently, our study showed that chronic treatment with LA effectively
inhibited tau hyperphosphorylation by alleviation of oxidative stress, inflammation,
and ferroptosis in the brains of an AD mouse model [309].
Surprisingly, the recent study found that Lf as an iron chelator and a therapeutic
agent has been investigated to identify its roles in AD. The affinity of Lf for iron is
300 times higher than that of TF. In weakly acidic environments, the affinity is further
enhanced, which may be related to the transfer of iron from transferrin to Lf during
inflammation [67]. Several experiments showed that Lf is expressed in many neurons
and in some astrocytes, microglia, and oligodendrocytes of the human brain [13, 147]
and that it could involve in aging processes [159, 226]. Further research findings sug-
gested that Lf is highly expressed in both extracellular SPs composed of aggregated
Aβ peptide and in intracellular NFTs composed of aggregated hyperphosphorylated
tau protein, which are suggesting the link of Lf and AD-like pathologies [147, 289].
Therefore, the use of exogenous Lf as a therapeutic agent has been investigated to
identify its roles in AD. Administrated Lf could stimulate the non-amyloidogenic
processing of APP and α-secretase catalytic activity and expression in AD model
mice and Lf enhanced the α-secretase-dependent APP process through the ERK1/2-
CREB and HIF-1α pathways in vitro and in vivo [116]. In combination with other
biological functions of Lf, such as its participation in metal ion metabolism and the
regulation of cell proliferation and apoptosis, we infer that Lf may be a potential
therapeutic agent to delay the pathological progress of AD.
90 T. Wang et al.
5.8 Conclusion
Why is it so hard to find a cure to AD? The main explanation is that the complex
causes of AD. It presents itself typically late in life and under the influence of many
combined genetic and lifestyle risk modifiers such as metabolic disorder of metal
ions, smoking, alcohol use, diabetes, body weight, hypertension, and physical and
intellectual activity levels, and perhaps even certain diets. Researchers have done
a lot of research on these pathogenic factors [103, 246, 282]. Recently, the study
of the pathogenesis of AD using iron as a target has become a research hotspot [8,
32, 56, 160, 161, 164, 176, 242]. The use of iron chelators to chelate excess iron
in the brains of AD patients has also become a new strategy for the treatment of
AD [78]. However, many therapeutic drugs for AD are still difficult to translate into
clinical application, largely due to the existence of BBB, which prevents drugs from
entering the brain to play a role. Therefore, effective drugs for AD must be easy to
pass through BBB. In our bodies, many proteins or molecules bind to iron to form
active complexes, such as LA and Lf. Some researchers have confirmed that treating
AD transgenic mice with LA and LF can alleviate their pathological symptoms [116,
309], but the specific mechanism of its action is still unclear, and further studies are
needed. Hopefully, in the meantime, Lf-based nano-drug molecules are also under
study. This drug molecule exhibits excellent biocompatibility and can easily cross
BBB to reach the lesion site, showing effective therapeutic effect. Future research
will also use iron as an opportunity to study the mechanism of the occurrence and
development of AD from the iron homeostasis to more fully explain the causes of
AD.
References
1. Abdalkader M, Lampinen R, Kanninen KM, Malm TM, Liddell JR (2018) Targeting Nrf2
to suppress ferroptosis and mitochondrial dysfunction in neurodegeneration. Front Neurosci
12:466
2. Adams TE, Mason AB, He QY, Halbrooks PJ, Briggs SK et al (2003) The position of arginine
124 controls the rate of iron release from the N-lobe of human serum transferrin. A structural
study. J Biol Chem 278:6027–6033
3. Agosta F, Pievani M, Sala S, Geroldi C, Galluzzi S et al (2011) White matter damage in
Alzheimer disease and its relationship to gray matter atrophy. Radiology 258:853–863
4. Ahmad KA, Ahmann JR, Migas MC, Waheed A, Britton RS et al (2002) Decreased liver
hepcidin expression in the Hfe knockout mouse. Blood Cells Mol Dis 29:361–366
5. Aisen P, Leibman A (1972) Lactoferrin and transferrin: a comparative study. Biochim Biophys
Acta 257:314–323
6. Ali-Rahmani F, Schengrund CL, Connor JR (2014) HFE gene variants, iron, and lipids: a
novel connection in Alzheimer’s disease. Front Pharmacol 5:165
7. Alimonti A, Ristori G, Giubilei F, Stazi MA, Pino A et al (2007) Serum chemical elements
and oxidative status in Alzheimer’s disease, Parkinson disease and multiple sclerosis. Neuro-
toxicology 28:450–456
8. Altamura S, Muckenthaler MU (2009) Iron toxicity in diseases of aging: Alzheimer’s disease,
Parkinson’s disease and atherosclerosis. J Alzheimers Dis 16:879–895
5 Iron Pathophysiology in Alzheimer’s Diseases 91
9. Alzheimer’s A (2016) 2016 Alzheimer’s disease facts and figures. Alzheimers Dement
12:459–509
10. Anderson ED, VanSlyke JK, Thulin CD, Jean F, Thomas G (1997) Activation of the furin
endoprotease is a multiple-step process: requirements for acidification and internal propeptide
cleavage. EMBO J 16:1508–1518
11. Andrasi E, Farkas E, Gawlik D, Rosick U, Bratter P (2000) Brain iron and zinc contents of
German patients with Alzheimer disease. J Alzheimers Dis 2:17–26
12. Arendt T (2009) Synaptic degeneration in Alzheimer’s disease. Acta Neuropathol
118:167–179
13. Arnold RR, Cole MF, McGhee JR (1977) A bactericidal effect for human lactoferrin. Science
197:263–265
14. Arosio P, Ingrassia R, Cavadini P (2009) Ferritins: a family of molecules for iron storage,
antioxidation and more. Biochim Biophys Acta 1790:589–599
15. Ayton S, Faux NG, Bush AI, Alzheimer’s Disease Neuroimaging I. (2015) Ferritin levels in
the cerebrospinal fluid predict Alzheimer’s disease outcomes and are regulated by APOE. Nat
Commun 6:6760
16. Ayton S, Fazlollahi A, Bourgeat P, Raniga P, Ng A et al (2017) Cerebral quantitative suscep-
tibility mapping predicts amyloid-β-related cognitive decline. Brain 140:2112–2119
17. Baker EN, Baker HM (2009) A structural framework for understanding the multifunctional
character of lactoferrin. Biochimie 91:3–10
18. Baker EN, Baker HM, Kidd RD (2002) Lactoferrin and transferrin: functional variations on
a common structural framework. Biochem Cell Biol 80:27–34
19. Bandyopadhyay S, Hartley DM, Cahill CM, Lahiri DK, Chattopadhyay N, Rogers JT (2006)
Interleukin-1alpha stimulates non-amyloidogenic pathway by alpha-secretase (ADAM-10 and
ADAM-17) cleavage of APP in human astrocytic cells involving p38 MAP kinase. J Neurosci
Res 84:106–118
20. Barnham KJ, Masters CL, Bush AI (2004) Neurodegenerative diseases and oxidative stress.
Nat Rev Drug Discov 3:205–214
21. Bartzokis G (2011) Alzheimer’s disease as homeostatic responses to age-related myelin break-
down. Neurobiol Aging 32:1341–1371
22. Bateman RJ, Aisen PS, De Strooper B, Fox NC, Lemere CA et al (2011) Autosomal-
dominant Alzheimer’s disease: a review and proposal for the prevention of Alzheimer’s dis-
ease. Alzheimers Res Ther 3:1
23. Baum L, Chan IH, Cheung SK, Goggins WB, Mok V et al (2010) Serum zinc is decreased in
Alzheimer’s disease and serum arsenic correlates positively with cognitive ability. Biometals
23:173–179
24. Belaidi AA, Bush AI (2016) Iron neurochemistry in Alzheimer’s disease and Parkinson’s
disease: targets for therapeutics. J Neurochem 139(Suppl 1):179–197
25. Berens C, Streicher B, Schroeder R, Hillen W (1998) Visualizing metal-ion-binding sites in
group I introns by iron(II)-mediated Fenton reactions. Chem Biol 5:163–175
26. Bhatt L, Murphy C, O’Driscoll LS, Carmo-Fonseca M, McCaffrey MW, Fleming JV (2010)
N-glycosylation is important for the correct intracellular localization of HFE and its ability
to decrease cell surface transferrin binding. FEBS J 277:3219–3234
27. Bocca B, Forte G, Petrucci F, Pino A, Marchione F et al (2005) Monitoring of chemical
elements and oxidative damage in patients affected by Alzheimer’s disease. Ann Ist Super
Sanita 41:197–203
28. Boddaert N, Le Quan Sang KH, Rotig A, Leroy-Willig A, Gallet S et al (2007) Selective iron
chelation in Friedreich ataxia: biologic and clinical implications. Blood 110:401–408
29. Borgna-Pignatti C, Marsella M (2015) Iron chelation in Thalassemia major. Clin Ther
37:2866–2877
30. Bostrom F, Hansson O, Gerhardsson L, Lundh T, Minthon L et al (2009) CSF Mg and Ca as
diagnostic markers for dementia with Lewy bodies. Neurobiol Aging 30:1265–1271
31. Bou-Abdallah F, Yang H, Awomolo A, Cooper B, Woodhall MR et al (2014) Functionality of
the three-site ferroxidase center of Escherichia coli bacterial ferritin (EcFtnA). Biochemistry
53:483–495
92 T. Wang et al.
32. Bousejra-ElGarah F, Bijani C, Coppel Y, Faller P, Hureau C (2011) Iron(II) binding to amyloid-
β, the Alzheimer’s peptide. Inorg Chem 50:9024–9030
33. Bradley-Whitman MA, Timmons MD, Beckett TL, Murphy MP, Lynn BC, Lovell MA (2014)
Nucleic acid oxidation: an early feature of Alzheimer’s disease. J Neurochem 128:294–304
34. Brickman AM, Provenzano FA, Muraskin J, Manly JJ, Blum S et al (2012) Regional white
matter hyperintensity volume, not hippocampal atrophy, predicts incident Alzheimer disease
in the community. Arch Neurol 69:1621–1627
35. Brickman AM, Zahodne LB, Guzman VA, Narkhede A, Meier IB et al (2015) Reconsidering
harbingers of dementia: progression of parietal lobe white matter hyperintensities predicts
Alzheimer’s disease incidence. Neurobiol Aging 36:27–32
36. Brown JP, Hewick RM, Hellstrom I, Hellstrom KE, Doolittle RF, Dreyer WJ (1982) Human
melanoma-associated antigen p97 is structurally and functionally related to transferrin. Nature
296:171–173
37. Burdo JR, Connor JR (2003) Brain iron uptake and homeostatic mechanisms: an overview.
Biometals 16:63–75
38. Burns A, Zaudig M (2002) Mild cognitive impairment in older people. Lancet 360:1963–1965
39. Bush AI (2003) The metallobiology of Alzheimer’s disease. Trends Neurosci 26:207–214
40. Butterfield DA, Drake J, Pocernich C, Castegna A (2001) Evidence of oxidative damage in
Alzheimer’s disease brain: central role for amyloid β-peptide. Trends Mol Med 7:548–554
41. Cai Z, Xiao M (2016) Oligodendrocytes and Alzheimer’s disease. Int J Neurosci 126:97–104
42. Cardozo-Pelaez F, Song S, Parthasarathy A, Hazzi C, Naidu K, Sanchez-Ramos J (1999)
Oxidative DNA damage in the aging mouse brain. Mov Disord 14:972–980
43. Carlson ES, Tkac I, Magid R, O’Connor MB, Andrews NC et al (2009) Iron is essential for
neuron development and memory function in mouse hippocampus. J Nutr 139:672–679
44. Carro E, Bartolome F, Bermejo-Pareja F, Villarejo-Galende A, Molina JA et al (2017) Early
diagnosis of mild cognitive impairment and Alzheimer’s disease based on salivary lactoferrin.
Alzheimers Dement (Amst) 8:131–138
45. Carvajal FJ, Mattison HA, Cerpa W (2016) Role of NMDA receptor-mediated glutamatergic
signaling in chronic and acute neuropathologies. Neural Plast 2016:2701526
46. Chakraborty S, Kaur S, Guha S, Batra SK (2012) The multifaceted roles of neutrophil
gelatinase associated lipocalin (NGAL) in inflammation and cancer. Biochim Biophys Acta
1826:129–169
47. Chan YK, Sung HK, Sweeney G (2015) Iron metabolism and regulation by neutrophil
gelatinase-associated lipocalin in cardiomyopathy. Clin Sci (Lond) 129:851–862
48. Cheah JH, Kim SF, Hester LD, Clancy KW, Patterson SE 3rd et al (2006) NMDA receptor-
nitric oxide transmission mediates neuronal iron homeostasis via the GTPase Dexras1. Neuron
51:431–440
49. Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F (2018) Oxidative
stress and the amyloid β peptide in Alzheimer’s disease. Redox Biol 14:450–564
50. Cho HH, Cahill CM, Vanderburg CR, Scherzer CR, Wang B et al (2010) Selective translational
control of the Alzheimer amyloid precursor protein transcript by iron regulatory protein-1. J
Biol Chem 285:31217–31232
51. Choi J, Lee HW, Suk K (2011) Increased plasma levels of lipocalin 2 in mild cognitive
impairment. J Neurol Sci 305:28–33
52. Chong MS, Sahadevan S (2005) Preclinical Alzheimer’s disease: diagnosis and prediction of
progression. Lancet Neurol 4:576–579
53. Connor JR, Menzies SL, St Martin SM, Mufson EJ (1990) Cellular distribution of transferrin,
ferritin, and iron in normal and aged human brains. J Neurosci Res 27:595–611
54. Connor JR, Menzies SL, St Martin SM, Mufson EJ (1992) A histochemical study of iron,
transferrin, and ferritin in Alzheimer’s diseased brains. J Neurosci Res 31:75–83
55. Connor JR, Snyder BS, Arosio P, Loeffler DA, LeWitt P (1995) A quantitative analysis
of isoferritins in select regions of aged, parkinsonian, and Alzheimer’s diseased brains. J
Neurochem 65:717–724
5 Iron Pathophysiology in Alzheimer’s Diseases 93
56. Connor JR, Snyder BS, Beard JL, Fine RE, Mufson EJ (1992) Regional distribution of iron
and iron-regulatory proteins in the brain in aging and Alzheimer’s disease. J Neurosci Res
31:327–335
57. Cooper LN, Bear MF (2012) The BCM theory of synapse modification at 30: interaction of
theory with experiment. Nat Rev Neurosci 13:798–810
58. Cooper-Knock J, Kirby J, Ferraiuolo L, Heath PR, Rattray M, Shaw PJ (2012) Gene expression
profiling in human neurodegenerative disease. Nat Rev Neurol 8:518–530
59. Cornett CR, Markesbery WR, Ehmann WD (1998) Imbalances of trace elements related to
oxidative damage in Alzheimer’s disease brain. Neurotoxicology 19:339–345
60. Corrigan FM, Reynolds GP, Ward NI (1993) Hippocampal tin, aluminum and zinc in
Alzheimer’s disease. Biometals 6:149–154
61. Corsi B, Cozzi A, Arosio P, Drysdale J, Santambrogio P et al (2002) Human mitochondrial
ferritin expressed in HeLa cells incorporates iron and affects cellular iron metabolism. J Biol
Chem 277:22430–22437
62. Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL et al (1991) Intramuscular
desferrioxamine in patients with Alzheimer’s disease. Lancet 337:1304–1308
63. Crespo AC, Silva B, Marques L, Marcelino E, Maruta C et al (2014) Genetic and biochemical
markers in patients with Alzheimer’s disease support a concerted systemic iron homeostasis
dysregulation. Neurobiol Aging 35:777–785
64. Cristovao JS, Santos R, Gomes CM (2016) Metals and neuronal metal binding proteins impli-
cated in Alzheimer’s disease. Oxid Med Cell Longev 2016:9812178
65. Dekens DW, Naude PJ, Engelborghs S, Vermeiren Y, Van Dam D et al (2017) Neutrophil
gelatinase-associated lipocalin and its receptors in Alzheimer’s disease (AD) brain regions:
differential findings in AD with and without depression. J Alzheimers Dis 55:763–776
66. Devireddy LR, Gazin C, Zhu X, Green MR (2005) A cell-surface receptor for lipocalin 24p3
selectively mediates apoptosis and iron uptake. Cell 123:1293–1305
67. Dhennin-Duthille I, Masson M, Damiens E, Fillebeen C, Spik G, Mazurier J (2000) Lactoferrin
upregulates the expression of CD4 antigen through the stimulation of the mitogen-activated
protein kinase in the human lymphoblastic T Jurkat cell line. J Cell Biochem 79:583–593
68. Ding B, Chen KM, Ling HW, Sun F, Li X et al (2009) Correlation of iron in the hippocampus
with MMSE in patients with Alzheimer’s disease. J Magn Reson Imaging 29:793–798
69. Ding Q, Dimayuga E, Markesbery WR, Keller JN (2004) Proteasome inhibition increases
DNA and RNA oxidation in astrocyte and neuron cultures. J Neurochem 91:1211–1218
70. Diouf I, Fazlollahi A, Bush AI, Ayton S, Alzheimer’s disease Neuroimaging I. (2019) Cere-
brospinal fluid ferritin levels predict brain hypometabolism in people with underlying β-
amyloid pathology. Neurobiol Dis 124:335–339
71. Dixit R, Ross JL, Goldman YE, Holzbaur EL (2008) Differential regulation of dynein and
kinesin motor proteins by tau. Science 319:1086–1089
72. Dixon SJ (2017) Ferroptosis: bug or feature? Immunol Rev 277:150–157
73. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM et al (2012) Ferroptosis: an
iron-dependent form of nonapoptotic cell death. Cell 149:1060–1072
74. De Domenico I, McVey Ward D, Kaplan J (2008) Regulation of iron acquisition and storage:
consequences for iron-linked disorders. Nat Rev Mol Cell Biol 9:72–81
75. Duce JA, Tsatsanis A, Cater MA, James SA, Robb E et al (2010) Iron-export ferroxidase
activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell
142:857–867
76. van Duijn S, Bulk M, van Duinen SG, Nabuurs RJA, van Buchem MA et al (2017) Cortical
iron reflects severity of Alzheimer’s disease. J Alzheimers Dis 60:1533–1545
77. Dunn LL, Sekyere EO, Suryo Rahmanto Y, Richardson DR (2006) The function of
melanotransferrin: a role in melanoma cell proliferation and tumorigenesis. Carcinogenesis
27:2157–2169
78. Dusek P, Schneider SA, Aaseth J (2016) Iron chelation in the treatment of neurodegenerative
diseases. J Trace Elem Med Biol 38:81–92
94 T. Wang et al.
79. Elfawy HA, Das B (2019) Crosstalk between mitochondrial dysfunction, oxidative stress,
and age related neurodegenerative disease: etiologies and therapeutic strategies. Life Sci
218:165–184
80. Esteves IM, Lopes-Aguiar C, Rossignoli MT, Ruggiero RN, Broggini ACS et al (2017)
Chronic nicotine attenuates behavioral and synaptic plasticity impairments in a streptozo-
tocin model of Alzheimer’s disease. Neuroscience 353:87–97
81. Estrada JA, Contreras I, Pliego-Rivero FB, Otero GA (2014) Molecular mechanisms of cog-
nitive impairment in iron deficiency: alterations in brain-derived neurotrophic factor and
insulin-like growth factor expression and function in the central nervous system. Nutr Neu-
rosci 17:193–206
82. Fava A, Pirritano D, Plastino M, Cristiano D, Puccio G et al (2013) The effect of lipoic acid
therapy on cognitive functioning in patients with Alzheimer’s disease. J Neurodegener Dis
2013:454253
83. Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA et al (1996) A novel MHC class
I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet 13:399–408
84. Feder JN, Penny DM, Irrinki A, Lee VK, Lebron JA et al (1998) The hemochromatosis gene
product complexes with the transferrin receptor and lowers its affinity for ligand binding. Proc
Natl Acad Sci U S A 95:1472–1477
85. Fischer FU, Wolf D, Scheurich A, Fellgiebel A, Alzheimer’s Disease Neuroimaging I. (2015)
Altered whole-brain white matter networks in preclinical Alzheimer’s disease. Neuroimage
Clin 8:660–666
86. Fonte J, Miklossy J, Atwood C, Martins R (2001) The severity of cortical Alzheimer’s type
changes is positively correlated with increased amyloid-β Levels: Resolubilization of amyloid-
β with transition metal ion chelators. J Alzheimers Dis 3:209–219
87. Food MR, Sekyere EO, Richardson DR (2002) The soluble form of the membrane-bound
transferrin homologue, melanotransferrin, inefficiently donates iron to cells via nonspecific
internalization and degradation of the protein. Eur J Biochem 269:4435–4445
88. Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC (2018) Neuronal cell death.
Physiol Rev 98:813–880
89. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA et al (2014) Inacti-
vation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol
16:1180–1191
90. Frisoni GB, Boccardi M, Barkhof F, Blennow K, Cappa S et al (2017) Strategic roadmap for
an early diagnosis of Alzheimer’s disease based on biomarkers. Lancet Neurol 16:661–676
91. Fujioka M, Taoka T, Matsuo Y, Mishima K, Ogoshi K et al (2003) Magnetic resonance imaging
shows delayed ischemic striatal neurodegeneration. Ann Neurol 54:732–747
92. Gaasch JA, Lockman PR, Geldenhuys WJ, Allen DD, Van der Schyf CJ (2007) Brain iron
toxicity: differential responses of astrocytes, neurons, and endothelial cells. Neurochem Res
32:1196–1208
93. Gabbita SP, Lovell MA, Markesbery WR (1998) Increased nuclear DNA oxidation in the
brain in Alzheimer’s disease. J Neurochem 71:2034–2040
94. Galasko DR, Peskind E, Clark CM, Quinn JF, Ringman JM et al (2012) Antioxidants for
Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures.
Arch Neurol 69:836–841
95. Galazka-Friedman J, Bauminger ER, Szlachta K, Friedman A (2012) The role of iron in neu-
rodegeneration–Mossbauer spectroscopy, electron microscopy, enzyme-linked immunosor-
bent assay and neuroimaging studies. J Phys: Condens Matter 24:244106
96. Gallagher JJ, Finnegan ME, Grehan B, Dobson J, Collingwood JF, Lynch MA (2012) Modest
amyloid deposition is associated with iron dysregulation, microglial activation, and oxidative
stress. J Alzheimers Dis 28:147–161
97. Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X (2016) Ferroptosis is an autophagic cell
death process. Cell Res 26:1021–1032
98. Gerhardsson L, Blennow K, Lundh T, Londos E, Minthon L (2009) Concentrations of metals,
β-amyloid and tau-markers in cerebrospinal fluid in patients with Alzheimer’s disease. Dement
Geriatr Cogn Disord 28:88–94
5 Iron Pathophysiology in Alzheimer’s Diseases 95
99. Gerstein M, Anderson BF, Norris GE, Baker EN, Lesk AM, Chothia C (1993) Domain clo-
sure in lactoferrin. Two hinges produce a see-saw motion between alternative close-packed
interfaces. J Mol Biol 234:357–372
100. Giambattistelli F, Bucossi S, Salustri C, Panetta V, Mariani S et al (2012) Effects of hemochro-
matosis and transferrin gene mutations on iron dyshomeostasis, liver dysfunction and on the
risk of Alzheimer’s disease. Neurobiol Aging 33:1633–1641
101. Giovannelli L, Decorosi F, Dolara P, Pulvirenti L (2003) Vulnerability to DNA damage in the
aging rat substantia nigra: a study with the comet assay. Brain Res 969:244–247
102. Gomes CM, Santos R (2013) Neurodegeneration in Friedreich’s ataxia: from defective frataxin
to oxidative stress. Oxid Med Cell Longev 2013:487534
103. Gomez-Pinilla F, Yang X (2018) System biology approach intersecting diet and cell
metabolism with pathogenesis of brain disorders. Prog Neurobiol 169:76–90
104. Gomme PT, McCann KB, Bertolini J (2005) Transferrin: structure, function and potential
therapeutic actions. Drug Discov Today 10:267–273
105. Gong NJ, Dibb R, Bulk M, van der Weerd L, Liu C (2019) Imaging β amyloid aggregation
and iron accumulation in Alzheimer’s disease using quantitative susceptibility mapping MRI.
Neuroimage 191:176–185
106. Gonzalez J, Jurado-Coronel JC, Avila MF, Sabogal A, Capani F, Barreto GE (2015) NMDARs
in neurological diseases: a potential therapeutic target. Int J Neurosci 125:315–327
107. Good PF, Perl DP, Bierer LM, Schmeidler J (1992) Selective accumulation of aluminum
and iron in the neurofibrillary tangles of Alzheimer’s disease: a laser microprobe (LAMMA)
study. Ann Neurol 31:286–292
108. Goodman L (1953) Alzheimer’s disease; a clinico-pathologic analysis of twenty-three cases
with a theory on pathogenesis. J Nerv Ment Dis 118:97–130
109. Goozee K, Chatterjee P, James I, Shen K, Sohrabi HR et al (2018) Elevated plasma ferritin
in elderly individuals with high neocortical amyloid-β load. Mol Psychiatry 23:1807–1812
110. Goralska M, Dackor R, Holley B, McGahan MC (2003) Alpha lipoic acid changes iron uptake
and storage in lens epithelial cells. Exp Eye Res 76:241–248
111. Goswami T, Andrews NC (2006) Hereditary hemochromatosis protein, HFE, interaction with
transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing. J Biol
Chem 281:28494–28498
112. Griffiths PD, Crossman AR (1993) Distribution of iron in the basal ganglia and neocortex in
postmortem tissue in Parkinson’s disease and Alzheimer’s disease. Dementia 4:61–65
113. Gu M, Owen AD, Toffa SE, Cooper JM, Dexter DT et al (1998) Mitochondrial function, GSH
and iron in neurodegeneration and Lewy body diseases. J Neurol Sci 158:24–29
114. Guo C, Wang T, Zheng W, Shan ZY, Teng WP, Wang ZY (2013) Intranasal deferoxamine
reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a trans-
genic mouse model of Alzheimer’s disease. Neurobiol Aging 34:562–575
115. Guo C, Wang P, Zhong ML, Wang T, Huang XS et al (2013) Deferoxamine inhibits iron
induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neu-
rochem Int 62:165–172
116. Guo C, Yang ZH, Zhang S, Chai R, Xue H et al (2017) Intranasal lactoferrin enhances
alpha-secretase-dependent amyloid precursor protein processing via the ERK1/2-CREB and
HIF-1alpha pathways in an Alzheimer’s disease mouse model. Neuropsychopharmacology
42:2504–2515
117. Guo C, Zhang YX, Wang T, Zhong ML, Yang ZH et al (2015) Intranasal deferoxamine atten-
uates synapse loss via up-regulating the P38/HIF-1alpha pathway on the brain of APP/PS1
transgenic mice. Front Aging Neurosci 7:104
118. Haeger P, Alvarez A, Leal N, Adasme T, Nunez MT, Hidalgo C (2010) Increased hippocampal
expression of the divalent metal transporter 1 (DMT1) mRNA variants 1B and +IRE and
DMT1 protein after NMDA-receptor stimulation or spatial memory training. Neurotox Res
17:238–247
119. Hallgren B, Sourander P (1958) The effect of age on the non-haemin iron in the human brain.
J Neurochem 3:41–51
96 T. Wang et al.
120. Hambright WS, Fonseca RS, Chen L, Na R, Ran Q (2017) Ablation of ferroptosis regulator
glutathione peroxidase 4 in forebrain neurons promotes cognitive impairment and neurode-
generation. Redox Biol 12:8–17
121. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM et al (2001) Does oxidative
damage to DNA increase with age? Proc Natl Acad Sci U S A 98:10469–10474
122. Han M, Kim J (2015) Effect of dietary iron loading on recognition memory in growing rats.
PLoS ONE 10:e0120609
123. Hare DJ, Doecke JD, Faux NG, Rembach A, Volitakis I et al (2015) Decreased plasma iron
in Alzheimer’s disease is due to transferrin desaturation. ACS Chem Neurosci 6:398–402
124. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F et al (2015) Neuroinflam-
mation in Alzheimer’s disease. Lancet Neurol 14:388–405
125. Hentze MW, Muckenthaler MU, Andrews NC (2004) Balancing acts: molecular control of
mammalian iron metabolism. Cell 117:285–297
126. Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: the role of inflammation in
Alzheimer disease. Nat Rev Neurosci 16:358–372
127. Hirano T, Yamaguchi R, Asami S, Iwamoto N, Kasai H (1996) 8-hydroxyguanine levels in
nuclear DNA and its repair activity in rat organs associated with age. J Gerontol A Biol Sci
Med Sci 51:B303–B307
128. Hofer T, Perry G (2016) Nucleic acid oxidative damage in Alzheimer’s disease-explained by
the hepcidin-ferroportin neuronal iron overload hypothesis? J Trace Elem Med Biol 38:1–9
129. Holland R, McIntosh AL, Finucane OM, Mela V, Rubio-Araiz A et al (2018) Inflammatory
microglia are glycolytic and iron retentive and typify the microglia in APP/PS1 mice. Brain
Behav Immun 68:183–196
130. Honda K, Smith MA, Zhu X, Baus D, Merrick WC et al (2005) Ribosomal RNA in Alzheimer
disease is oxidized by bound redox-active iron. J Biol Chem 280:20978–20986
131. Horowitz MP, Greenamyre JT (2010) Mitochondrial iron metabolism and its role in neurode-
generation. J Alzheimers Dis 20(Suppl 2):S551–S568
132. House E, Collingwood J, Khan A, Korchazkina O, Berthon G, Exley C (2004) Aluminium,
iron, zinc and copper influence the in vitro formation of amyloid fibrils of Aβ42 in a man-
ner which may have consequences for metal chelation therapy in Alzheimer’s disease. J
Alzheimers Dis 6:291–301
133. House MJ, St Pierre TG, McLean C (2008) 1.4T study of proton magnetic relaxation rates,
iron concentrations, and plaque burden in Alzheimer’s disease and control postmortem brain
tissue. Magn Reson Med 60:41–52
134. Hozumi I, Hasegawa T, Honda A, Ozawa K, Hayashi Y et al (2011) Patterns of levels of
biological metals in CSF differ among neurodegenerative diseases. J Neurol Sci 303:95–99
135. Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE et al (1999) The Aβ peptide
of Alzheimer’s disease directly produces hydrogen peroxide through metal ion reduction.
Biochemistry 38:7609–7616
136. Huang CW, Wang SJ, Wu SJ, Yang CC, Huang MW et al (2013) Potential blood biomarker
for disease severity in the Taiwanese population with Alzheimer’s disease. Am J Alzheimers
Dis Other Demen 28:75–83
137. Hui Y, Wang D, Li W, Zhang L, Jin J et al (2011) Long-term overexpression of heme oxygenase
1 promotes tau aggregation in mouse brain by inducing tau phosphorylation. J Alzheimers
Dis 26:299–313
138. Illing AC, Shawki A, Cunningham CL, Mackenzie B (2012) Substrate profile and metal-ion
selectivity of human divalent metal-ion transporter-1. J Biol Chem 287:30485–30496
139. Ising C, Stanley M, Holtzman DM (2015) Current thinking on the mechanistic basis of
Alzheimer’s and implications for drug development. Clin Pharmacol Ther 98:469–471
140. Jiang T, Sun Q, Chen S (2016) Oxidative stress: A major pathogenesis and potential therapeutic
target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Prog Neurobiol
147:1–19
141. Juurlink BH, Thorburne SK, Hertz L (1998) Peroxide-scavenging deficit underlies oligoden-
drocyte susceptibility to oxidative stress. Glia 22:371–378
5 Iron Pathophysiology in Alzheimer’s Diseases 97
164. Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C (2018) Iron and Alzheimer’s disease: from
pathogenesis to therapeutic implications. Front Neurosci 12:632
165. Liu Y, Lee SY, Neely E, Nandar W, Moyo M et al (2011) Mutant HFE H63D protein is
associated with prolonged endoplasmic reticulum stress and increased neuronal vulnerability.
J Biol Chem 286:13161–13170
166. Liu B, Moloney A, Meehan S, Morris K, Thomas SE et al (2011) Iron promotes the toxicity
of amyloid β peptide by impeding its ordered aggregation. J Biol Chem 286:4248–4256
167. Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J et al (2017) Dementia
prevention, intervention, and care. Lancet 390:2673–2734
168. Loboda A, Jazwa A, Grochot-Przeczek A, Rutkowski AJ, Cisowski J et al (2008) Heme
oxygenase-1 and the vascular bed: from molecular mechanisms to therapeutic opportunities.
Antioxid Redox Signal 10:1767–1812
169. Lopes KO, Sparks DL, Streit WJ (2008) Microglial dystrophy in the aged and Alzheimer’s
disease brain is associated with ferritin immunoreactivity. Glia 56:1048–1060
170. Lovell MA, Gabbita SP, Markesbery WR (1999) Increased DNA oxidation and decreased
levels of repair products in Alzheimer’s disease ventricular CSF. J Neurochem 72:771–776
171. Lu T, Pan Y, Kao SY, Li C, Kohane I et al (2004) Gene regulation and DNA damage in the
ageing human brain. Nature 429:883–891
172. Lu LN, Qian ZM, Wu KC, Yung WH, Ke Y (2017) Expression of iron transporters and
pathological hallmarks of Parkinson’s and Alzheimer’s diseases in the brain of young, adult,
and aged rats. Mol Neurobiol 54:5213–5224
173. Maamoun H, Benameur T, Pintus G, Munusamy S, Agouni A (2019) Crosstalk between
oxidative stress and endoplasmic reticulum (ER) stress in endothelial dysfunction and aberrant
angiogenesis associated with diabetes: a focus on the protective roles of heme oxygenase
(HO)-1. Front Physiol 10:70
174. Magnani E, Fan J, Gasparini L, Golding M, Williams M et al (2007) Interaction of tau protein
with the dynactin complex. EMBO J 26:4546–4554
175. Mairuae N, Hall Ii EC, Cheepsunthorn P, Lee SY, Connor JR (2010) The H63D HFE gene
variant promotes activation of the intrinsic apoptotic pathway via mitochondria dysfunction
following β-amyloid peptide exposure. J Neurosci Res 88:3079–3089
176. Mandel S, Amit T, Bar-Am O, Youdim MB (2007) Iron dysregulation in Alzheimer’s disease:
multimodal brain permeable iron chelating drugs, possessing neuroprotective-neurorescue
and amyloid precursor protein-processing regulatory activities as therapeutic agents. Prog
Neurobiol 82:348–360
177. Marro S, Chiabrando D, Messana E, Stolte J, Turco E et al (2010) Heme controls ferroportin1
(FPN1) transcription involving Bach1, Nrf2 and a MARE/ARE sequence motif at position—
7007 of the FPN1 promoter. Haematologica 95:1261–1268
178. Mattson MP (2004) Metal-catalyzed disruption of membrane protein and lipid signaling in
the pathogenesis of neurodegenerative disorders. Ann N Y Acad Sci 1012:37–50
179. Matute C (2010) Calcium dyshomeostasis in white matter pathology. Cell Calcium
47:150–157
180. McCarthy RC, Sosa JC, Gardeck AM, Baez AS, Lee CH, Wessling-Resnick M (2018)
Inflammation-induced iron transport and metabolism by brain microglia. J Biol Chem
293:7853–7863
181. McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, Sager G et al (2001) An iron-regulated
ferric reductase associated with the absorption of dietary iron. Science 291:1755–1759
182. Meadowcroft MD, Connor JR, Smith MB, Yang QX (2009) MRI and histological analysis of
β-amyloid plaques in both human Alzheimer’s disease and APP/PS1 transgenic mice. J Magn
Reson Imaging: JMRI 29:997–1007
183. Meadowcroft MD, Connor JR, Yang QX (2015) Cortical iron regulation and inflammatory
response in Alzheimer’s disease and APPSWE/PS1DeltaE9 mice: a histological perspective.
Front Neurosci 9:255
184. Mecocci P, MacGarvey U, Beal MF (1994) Oxidative damage to mitochondrial DNA is
increased in Alzheimer’s disease. Ann Neurol 36:747–751
5 Iron Pathophysiology in Alzheimer’s Diseases 99
185. Mecocci P, Polidori MC, Ingegni T, Cherubini A, Chionne F et al (1998) Oxidative damage
to DNA in lymphocytes from AD patients. Neurology 51:1014–1017
186. Mehlenbacher M, Poli M, Arosio P, Santambrogio P, Levi S et al (2017) Iron oxidation and
core formation in recombinant heteropolymeric human ferritins. Biochemistry 56:3900–3912
187. Mena NP, Urrutia PJ, Lourido F, Carrasco CM, Nunez MT (2015) Mitochondrial iron home-
ostasis and its dysfunctions in neurodegenerative disorders. Mitochondrion 21:92–105
188. Di Meo I, Tiranti V (2018) Classification and molecular pathogenesis of NBIA syndromes.
Eur J Paediatr Neurol 22:272–284
189. Mesquita SD, Ferreira AC, Falcao AM, Sousa JC, Oliveira TG et al (2014) Lipocalin 2
modulates the cellular response to amyloid β. Cell Death Differ 21:1588–1599
190. Migliore L, Fontana I, Trippi F, Colognato R, Coppede F et al (2005) Oxidative DNA damage
in peripheral leukocytes of mild cognitive impairment and AD patients. Neurobiol Aging
26:567–573
191. Miyamoto T, Kashima H, Suzuki A, Kikuchi N, Konishi I et al (2011) Laser-captured
microdissection-microarray analysis of the genes involved in endometrial carcinogenesis:
stepwise up-regulation of lipocalin2 expression in normal and neoplastic endometria and its
functional relevance. Hum Pathol 42:1265–1274
192. Molina JA, Jimenez-Jimenez FJ, Aguilar MV, Meseguer I, Mateos-Vega CJ et al (1998)
Cerebrospinal fluid levels of transition metals in patients with Alzheimer’s disease. J Neural
Transm (Vienna) 105:479–488
193. Mouton-Liger F, Paquet C, Dumurgier J, Bouras C, Pradier L et al (2012) Oxidative stress
increases BACE1 protein levels through activation of the PKR-eIF2alpha pathway. Biochim
Biophys Acta 1822:885–896
194. Multhaup G, Schlicksupp A, Hesse L, Beher D, Ruppert T et al (1996) The amyloid pre-
cursor protein of Alzheimer’s disease in the reduction of copper(II) to copper(I). Science
271:1406–1409
195. Munoz P, Humeres A (2012) Iron deficiency on neuronal function. Biometals 25:825–835
196. Munoz P, Humeres A, Elgueta C, Kirkwood A, Hidalgo C, Nunez MT (2011) Iron medi-
ates N-methyl-D-aspartate receptor-dependent stimulation of calcium-induced pathways and
hippocampal synaptic plasticity. J Biol Chem 286:13382–13392
197. Nakamichi N, Oikawa H, Kambe Y, Yoneda Y (2004) Relevant modulation by ferrous ions of
N-methyl-D-aspartate receptors in ischemic brain injuries. Curr Neurovasc Res 1:429–440
198. Nandar W, Connor JR (2011) HFE gene variants affect iron in the brain. J Nutr 141:729S–739S
199. Nasrabady SE, Rizvi B, Goldman JE, Brickman AM (2018) White matter changes in
Alzheimer’s disease: a focus on myelin and oligodendrocytes. Acta Neuropathol Commun
6:22
200. Naude PJ, Nyakas C, Eiden LE, Ait-Ali D, van der Heide R et al (2012) Lipocalin 2: novel
component of proinflammatory signaling in Alzheimer’s disease. FASEB J 26:2811–2823
201. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A et al (2004) Hepcidin regu-
lates cellular iron efflux by binding to ferroportin and inducing its internalization. Science
306:2090–2093
202. Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B et al (2001) Lack of hepcidin
gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2)
knockout mice. Proc Natl Acad Sci U S A 98:8780–8785
203. Nicolas G, Bennoun M, Porteu A, Mativet S, Beaumont C et al (2002) Severe iron deficiency
anemia in transgenic mice expressing liver hepcidin. Proc Natl Acad Sci U S A 99:4596–4601
204. Nicolas G, Viatte L, Lou DQ, Bennoun M, Beaumont C et al (2003) Constitutive hep-
cidin expression prevents iron overload in a mouse model of hemochromatosis. Nat Genet
34:97–101
205. Niki E, Yoshida Y, Saito Y, Noguchi N (2005) Lipid peroxidation: mechanisms, inhibition,
and biological effects. Biochem Biophys Res Commun 338:668–676
206. Nnah IC, Wessling-Resnick M (2018) Brain iron homeostasis: a focus on microglial iron.
Pharmaceuticals (Basel) 11
100 T. Wang et al.
207. Nunomura A, Chiba S, Lippa CF, Cras P, Kalaria RN et al (2004) Neuronal RNA oxidation
is a prominent feature of familial Alzheimer’s disease. Neurobiol Dis 17:108–113
208. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A et al (2001) Oxidative damage is the
earliest event in Alzheimer disease. J Neuropathol Exp Neurol 60:759–767
209. Nunomura A, Perry G, Pappolla MA, Friedland RP, Hirai K et al (2000) Neuronal oxida-
tive stress precedes amyloid-β deposition in down syndrome. J Neuropathol Exp Neurol
59:1011–1017
210. Nunomura A, Perry G, Pappolla MA, Wade R, Hirai K et al (1999) RNA oxidation is a
prominent feature of vulnerable neurons in Alzheimer’s disease. J Neurosci 19:1959–1964
211. Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A et al (2005) Identification
of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells.
Nat Genet 37:1264–1269
212. Ou P, Tritschler HJ, Wolff SP (1995) Thioctic (lipoic) acid: a therapeutic metal-chelating
antioxidant? Biochem Pharmacol 50:123–126
213. Ozcankaya R, Delibas N (2002) Malondialdehyde, superoxide dismutase, melatonin, iron,
copper, and zinc blood concentrations in patients with Alzheimer disease: cross-sectional
study. Croat Med J 43:28–32
214. Park CH, Valore EV, Waring AJ, Ganz T (2001) Hepcidin, a urinary antimicrobial peptide
synthesized in the liver. J Biol Chem 276:7806–7810
215. Paul S, Connor JA (2010) NR2B-NMDA receptor-mediated increases in intracellular Ca2+
concentration regulate the tyrosine phosphatase, STEP, and ERK MAP kinase signaling. J
Neurochem 114:1107–1118
216. Pelizzoni I, Macco R, Morini MF, Zacchetti D, Grohovaz F, Codazzi F (2011) Iron handling
in hippocampal neurons: activity-dependent iron entry and mitochondria-mediated neurotox-
icity. Aging Cell 10:172–183
217. Perez DR, Sklar LA, Chigaev A (2019) Clioquinol: to harm or heal. Pharmacol Ther
218. Persson T, Popescu BO, Cedazo-Minguez A (2014) Oxidative stress in Alzheimer’s disease:
why did antioxidant therapy fail? Oxid Med Cell Longev 2014:427318
219. Pierrel F, Cobine PA, Winge DR (2007) Metal Ion availability in mitochondria. Biometals
20:675–682
220. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B et al (2001) A new mouse liver-specific
gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overex-
pressed during iron overload. J Biol Chem 276:7811–7819
221. Pini L, Pievani M, Bocchetta M, Altomare D, Bosco P et al (2016) Brain atrophy in Alzheimer’s
disease and aging. Ageing Res Rev 30:25–48
222. Pooler AM, Noble W, Hanger DP (2014) A role for tau at the synapse in Alzheimer’s disease
pathogenesis. Neuropharmacology 76 Pt A:1–8
223. Pratico D, Uryu K, Leight S, Trojanoswki JQ, Lee VM (2001) Increased lipid peroxidation
precedes amyloid plaque formation in an animal model of Alzheimer amyloidosis. J Neurosci
21:4183–4187
224. Puzzo D (2019) Abeta oligomers: role at the synapse. Aging (Albany NY) 11:1077–1078
225. Qian ZM, Shen X (2001) Brain iron transport and neurodegeneration. Trends Mol Med
7:103–108
226. Qian ZM, Wang Q (1998) Expression of iron transport proteins and excessive iron accumu-
lation in the brain in neurodegenerative disorders. Brain Res Brain Res Rev 27:257–267
227. Radi E, Formichi P, Battisti C, Federico A (2014) Apoptosis and oxidative stress in neurode-
generative diseases. J Alzheimers Dis 42(Suppl 3):S125–S152
228. Radlowski EC, Johnson RW (2013) Perinatal iron deficiency and neurocognitive development.
Front Hum Neurosci 7:585
229. Raha AA, Vaishnav RA, Friedland RP, Bomford A, Raha-Chowdhury R (2013) The systemic
iron-regulatory proteins hepcidin and ferroportin are reduced in the brain in Alzheimer’s
disease. Acta Neuropathol Commun 1:55
230. Rajan MT, Jagannatha Rao KS, Mamatha BM, Rao RV, Shanmugavelu P et al (1997) Quan-
tification of trace elements in normal human brain by inductively coupled plasma atomic
emission spectrometry. J Neurol Sci 146:153–166
5 Iron Pathophysiology in Alzheimer’s Diseases 101
231. Rakba N, Aouad F, Henry C, Caris C, Morel I et al (1998) Iron mobilisation and cellular
protection by a new synthetic chelator O-Trensox. Biochem Pharmacol 55:1797–1806
232. Ramos P, Santos A, Pinto NR, Mendes R, Magalhaes T, Almeida A (2014) Iron levels in the
human brain: a post-mortem study of anatomical region differences and age-related changes.
J Trace Elem Med Biol 28:13–17
233. Raven EP, Lu PH, Tishler TA, Heydari P, Bartzokis G (2013) Increased iron levels and
decreased tissue integrity in hippocampus of Alzheimer’s disease detected in vivo with mag-
netic resonance imaging. J Alzheimers Dis 37:127–136
234. Richardson DR (2000) The role of the membrane-bound tumour antigen, melanotransferrin
(p97), in iron uptake by the human malignant melanoma cell. Eur J Biochem 267:1290–1298
235. Rishi G, Wallace DF, Subramaniam VN (2015) Hepcidin: regulation of the master iron regu-
lator. Biosci Rep 35
236. Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M et al (2003) Metal-protein
attenuation with iodochlorhydroxyquin (clioquinol) targeting Aβ amyloid deposition and
toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch Neurol 60:1685–1691
237. Robert A, Liu Y, Nguyen M, Meunier B (2015) Regulation of copper and iron homeosta-
sis by metal chelators: a possible chemotherapy for Alzheimer’s disease. Acc Chem Res
48:1332–1339
238. Rogers JT, Bush AI, Cho HH, Smith DH, Thomson AM et al (2008) Iron and the translation
of the amyloid precursor protein (APP) and ferritin mRNAs: riboregulation against neural
oxidative damage in Alzheimer’s disease. Biochem Soc Trans 36:1282–1287
239. Roher AE, Weiss N, Kokjohn TA, Kuo YM, Kalback W et al (2002) Increased Aβ peptides and
reduced cholesterol and myelin proteins characterize white matter degeneration in Alzheimer’s
disease. Biochemistry 41:11080–11090
240. Rottkamp CA, Raina AK, Zhu X, Gaier E, Bush AI et al (2001) Redox-active iron mediates
amyloid-β toxicity. Free Radic Biol Med 30:447–450
241. Sabbir MG (2018) Loss of Ca(2+) /calmodulin dependent protein kinase kinase 2 leads to
aberrant transferrin phosphorylation and trafficking: a potential biomarker for Alzheimer’s
disease. Front Mol Biosci 5:99
242. Salvador GA (2010) Iron in neuronal function and dysfunction. BioFactors 36:103–110
243. Salvador GA, Uranga RM, Giusto NM (2010) Iron and mechanisms of neurotoxicity. Int J
Alzheimers Dis 2011:720658
244. Santambrogio P, Biasiotto G, Sanvito F, Olivieri S, Arosio P, Levi S (2007) Mitochondrial
ferritin expression in adult mouse tissues. J Histochem Cytochem 55:1129–1137
245. Sawikr Y, Yarla NS, Peluso I, Kamal MA, Aliev G, Bishayee A (2017) Neuroinflammation in
Alzheimer’s disease: the preventive and therapeutic potential of polyphenolic nutraceuticals.
Adv Protein Chem Struct Biol 108:33–57
246. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB et al (2016) Alzheimer’s
disease. Lancet 388:505–517
247. Schipper HM, Song W (2015) A heme oxygenase-1 transducer model of degenerative and
developmental brain disorders. Int J Mol Sci 16:5400–5419
248. Schipper HM, Song W, Tavitian A, Cressatti M (2019) The sinister face of heme oxygenase-1
in brain aging and disease. Prog Neurobiol 172:40–70
249. Schipper HM, Song W, Zukor H, Hascalovici JR, Zeligman D (2009) Heme oxygenase-1 and
neurodegeneration: expanding frontiers of engagement. J Neurochem 110:469–485
250. Schneider C, Pratt DA, Porter NA, Brash AR (2007) Control of oxygenation in lipoxygenase
and cyclooxygenase catalysis. Chem Biol 14:473–488
251. Schon EA, Area-Gomez E (2013) Mitochondria-associated ER membranes in Alzheimer
disease. Mol Cell Neurosci 55:26–36
252. Schrag M, Mueller C, Oyoyo U, Smith MA, Kirsch WM (2011) Iron, zinc and copper in
the Alzheimer’s disease brain: a quantitative meta-analysis. Some insight on the influence of
citation bias on scientific opinion. Prog Neurobiol 94:296–306
253. Seiler A, Schneider M, Forster H, Roth S, Wirth EK et al (2008) Glutathione peroxidase 4
senses and translates oxidative stress into 12/15-lipoxygenase dependent—and AIF-mediated
cell death. Cell Metab 8:237–248
102 T. Wang et al.
254. Sekyere EO, Dunn LL, Suryo Rahmanto Y, Richardson DR (2006) Role of melanotransferrin
in iron metabolism: studies using targeted gene disruption in vivo. Blood 107:2599–2601
255. Shan X, Lin CL (2006) Quantification of oxidized RNAs in Alzheimer’s disease. Neurobiol
Aging 27:657–662
256. Shichiri M (2014) The role of lipid peroxidation in neurological disorders. J Clin Biochem
Nutr 54:151–160
257. Shimada A, Keino H, Satoh M, Kishikawa M, Hosokawa M (2003) Age-related loss of
synapses in the frontal cortex of SAMP10 mouse: a model of cerebral degeneration. Synapse
48:198–204
258. Shin RW, Kruck TP, Murayama H, Kitamoto T (2003) A novel trivalent cation chelator
Feralex dissociates binding of aluminum and iron associated with hyperphosphorylated tau
of Alzheimer’s disease. Brain Res 961:139–146
259. da Silva VK, de Freitas BS, da Silva Dornelles A, Nery LR, Falavigna L et al (2014) Cannabid-
iol normalizes caspase 3, synaptophysin, and mitochondrial fission protein DNM1L expres-
sion levels in rats with brain iron overload: implications for neuroprotection. Mol Neurobiol
49:222–233
260. Smith MA, Harris PL, Sayre LM, Perry G (1997) Iron accumulation in Alzheimer disease is
a source of redox-generated free radicals. Proc Natl Acad Sci U S A 94:9866–9868
261. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S et al (2011) Toward defining the
preclinical stages of Alzheimer’s disease: recommendations from the National Institute on
Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease.
Alzheimers Dement 7:280–292
262. Spires-Jones TL, Hyman BT (2014) The intersection of amyloid β and tau at synapses in
Alzheimer’s disease. Neuron 82:756–771
263. Squitti R, Ghidoni R, Scrascia F, Benussi L, Panetta V et al (2011) Free copper distin-
guishes mild cognitive impairment subjects from healthy elderly individuals. J Alzheimers
Dis 23:239–248
264. Squitti R, Lupoi D, Pasqualetti P, Dal Forno G, Vernieri F et al (2002) Elevation of serum
copper levels in Alzheimer’s disease. Neurology 59:1153–1161
265. Squitti R, Salustri C, Siotto M, Ventriglia M, Vernieri F et al (2010) Ceruloplasmin/Transferrin
ratio changes in Alzheimer’s disease. Int J Alzheimers Dis 2011:231595
266. Srivastava RA, Jain JC (2002) Scavenger receptor class B type I expression and elemental
analysis in cerebellum and parietal cortex regions of the Alzheimer’s disease brain. J Neurol
Sci 196:45–52
267. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M et al (2017) Ferroptosis: a
regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171:273–285
268. Tang J, Oliveros A, Jang MH (2019) Dysfunctional mitochondrial bioenergetics and synaptic
degeneration in Alzheimer disease. Int Neurourol J 23:S5–S10
269. Telling ND, Everett J, Collingwood JF, Dobson J, van der Laan G et al (2017) Iron biochemistry
is correlated with amyloid plaque morphology in an established mouse model of Alzheimer’s
disease. Cell Chem Biol 24(1205–15):e3
270. Theil EC (2013) Ferritin: the protein nanocage and iron biomineral in health and in disease.
Inorg Chem 52:12223–12233
271. Thomas G (2002) Furin at the cutting edge: from protein traffic to embryogenesis and disease.
Nat Rev Mol Cell Biol 3:753–766
272. Thomsen MS, Andersen MV, Christoffersen PR, Jensen MD, Lichota J, Moos T (2015)
Neurodegeneration with inflammation is accompanied by accumulation of iron and ferritin
in microglia and neurons. Neurobiol Dis 81:108–118
273. Thorburne SK, Juurlink BH (1996) Low glutathione and high iron govern the susceptibility
of oligodendroglial precursors to oxidative stress. J Neurochem 67:1014–1022
274. Tiwari S, Ndisang JF (2014) The heme oxygenase system and type-1 diabetes. Curr Pharm
Des 20:1328–1337
275. Torsdottir G, Kristinsson J, Snaedal J, Johannesson T (2011) Ceruloplasmin and iron proteins
in the serum of patients with Alzheimer’s disease. Dement Geriatr Cogn Dis Extra 1:366–371
5 Iron Pathophysiology in Alzheimer’s Diseases 103
276. Tse KH, Herrup K (2017) DNA damage in the oligodendrocyte lineage and its role in brain
aging. Mech Ageing Dev 161:37–50
277. Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP et al (2013) Inflammation alters the
expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous
system cells. J Neurochem 126:541–549
278. Urrutia PJ, Mena NP, Nunez MT (2014) The interplay between iron accumulation, mitochon-
drial dysfunction, and inflammation during the execution step of neurodegenerative disorders.
Front Pharmacol 5:38
279. Ursini F, Bindoli A (1987) The role of selenium peroxidases in the protection against oxidative
damage of membranes. Chem Phys Lipids 44:255–276
280. Valore EV, Ganz T (2008) Posttranslational processing of hepcidin in human hepatocytes is
mediated by the prohormone convertase furin. Blood Cells Mol Dis 40:132–138
281. Varadarajan S, Yatin S, Aksenova M, Butterfield DA (2000) Review: Alzheimer’s amyloid
β-peptide-associated free radical oxidative stress and neurotoxicity. J Struct Biol 130:184–208
282. Veitch DP, Weiner MW, Aisen PS, Beckett LA, Cairns NJ et al (2019) Understanding disease
progression and improving Alzheimer’s disease clinical trials: recent highlights from the
Alzheimer’s disease neuroimaging initiative. Alzheimers Dement 15:106–152
283. Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS et al (2015) Prefibrillar Tau
oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo.
Neurobiol Dis 82:540–551
284. Vural H, Demirin H, Kara Y, Eren I, Delibas N (2010) Alterations of plasma magnesium, cop-
per, zinc, iron and selenium concentrations and some related erythrocyte antioxidant enzyme
activities in patients with Alzheimer’s disease. J Trace Elem Med Biol 24:169–173
285. Wally J, Halbrooks PJ, Vonrhein C, Rould MA, Everse SJ et al (2006) The crystal structure
of iron-free human serum transferrin provides insight into inter-lobe communication and
receptor binding. J Biol Chem 281:24934–24944
286. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R et al (2002) Naturally secreted
oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo.
Nature 416:535–539
287. Wan L, Nie G, Zhang J, Zhao B (2012) Overexpression of human wild-type amyloid-β protein
precursor decreases the iron content and increases the oxidative stress of neuroblastoma SH-
SY5Y cells. J Alzheimers Dis 30:523–530
288. Wang J, Markesbery WR, Lovell MA (2006) Increased oxidative damage in nuclear and
mitochondrial DNA in mild cognitive impairment. J Neurochem 96:825–832
289. Wang L, Sato H, Zhao S, Tooyama I (2010) Deposition of lactoferrin in fibrillar-type senile
plaques in the brains of transgenic mouse models of Alzheimer’s disease. Neurosci Lett
481:164–167
290. Wang J, Song N, Jiang H, Wang J, Xie J (2013) Pro-inflammatory cytokines modulate iron reg-
ulatory protein 1 expression and iron transportation through reactive oxygen/nitrogen species
production in ventral mesencephalic neurons. Biochim Biophys Acta 1832:618–625
291. Wang X, Wang W, Li L, Perry G, Lee HG, Zhu X (2014) Oxidative stress and mitochondrial
dysfunction in Alzheimer’s disease. Biochim Biophys Acta 1842:1240–1247
292. Wang P, Wu Q, Wu W, Li H, Guo Y et al (2017) Mitochondrial ferritin deletion exacerbates
β-amyloid-induced neurotoxicity in mice. Oxid Med Cell Longev 2017:1020357
293. Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA (2005) Increased oxidative damage in
nuclear and mitochondrial DNA in Alzheimer’s disease. J Neurochem 93:953–962
294. Ward A, Hudson JW (2014) p53-Dependent and cell specific epigenetic regulation of the
polo-like kinases under oxidative stress. PLoS ONE 9:e87918
295. Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L (2014) The role of iron in brain ageing
and neurodegenerative disorders. Lancet Neurol 13:1045–1060
296. White RS, Bhattacharya AK, Chen Y, Byrd M, McMullen MF et al (2016) Lysosomal iron
modulates NMDA receptor-mediated excitation via small GTPase, Dexras1. Mol Brain 9:38
297. Wimo A, Guerchet M, Ali GC, Wu YT, Prina AM et al (2017) The worldwide costs of dementia
2015 and comparisons with 2010. Alzheimers Dement 13:1–7
104 T. Wang et al.
298. Wong BX, Duce JA (2014) The iron regulatory capability of the major protein participants in
prevalent neurodegenerative disorders. Front Pharmacol 5:81
299. Wong BX, Tsatsanis A, Lim LQ, Adlard PA, Bush AI, Duce JA (2014) β-Amyloid precursor
protein does not possess ferroxidase activity but does stabilize the cell surface ferrous iron
exporter ferroportin. PLoS ONE 9:e114174
300. Wu WS, Zhao YS, Shi ZH, Chang SY, Nie GJ et al (2013) Mitochondrial ferritin attenuates β-
amyloid-induced neurotoxicity: reduction in oxidative damage through the Erk/P38 mitogen-
activated protein kinase pathways. Antioxid Redox Signal 18:158–169
301. Xu H, Jiang H, Xie J (2017) New insights into the crosstalk between NMDARs and iron:
implications for understanding pathology of neurological diseases. Front Mol Neurosci 10:71
302. Xu H, Wang Y, Song N, Wang J, Jiang H, Xie J (2017) New progress on the role of glia in
iron metabolism and iron-induced degeneration of dopamine neurons in Parkinson’s disease.
Front Mol Neurosci 10:455
303. Yamamoto A, Shin RW, Hasegawa K, Naiki H, Sato H et al (2002) Iron (III) induces aggre-
gation of hyperphosphorylated tau and its reduction to iron (II) reverses the aggregation:
implications in the formation of neurofibrillary tangles of Alzheimer’s disease. J Neurochem
82:1137–1147
304. Yang H, Guan H, Yang M, Liu Z, Takeuchi S et al (2015) Upregulation of mitochondrial ferritin
by proinflammatory cytokines: implications for a role in Alzheimer’s disease. J Alzheimers
Dis 45:797–811
305. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R et al (2014) Regulation of
ferroptotic cancer cell death by GPX4. Cell 156:317–331
306. You LH, Yan CZ, Zheng BJ, Ci YZ, Chang SY et al (2017) Astrocyte hepcidin is a key factor
in LPS-induced neuronal apoptosis. Cell Death Dis 8:e2676
307. Zatta P, Drago D, Bolognin S, Sensi SL (2009) Alzheimer’s disease, metal ions and metal
homeostatic therapy. Trends Pharmacol Sci 30:346–355
308. Zecca L, Stroppolo A, Gatti A, Tampellini D, Toscani M et al (2004) The role of iron and
copper molecules in the neuronal vulnerability of locus coeruleus and substantia nigra during
aging. Proc Natl Acad Sci U S A 101:9843–9848
309. Zhang YH, Wang DW, Xu SF, Zhang S, Fan YG et al (2018) alpha-Lipoic acid improves
abnormal behavior by mitigation of oxidative stress, inflammation, ferroptosis, and tauopathy
in P301S Tau transgenic mice. Redox Biol 14:535–548
310. Zilka N, Ferencik M, Hulin I (2006) Neuroinflammation in Alzheimer’s disease: protector or
promoter? Bratisl Lek Listy 107:374–383
311. Zmijewski JW, Landar A, Watanabe N, Dickinson DA, Noguchi N, Darley-Usmar VM (2005)
Cell signalling by oxidized lipids and the role of reactive oxygen species in the endothelium.
Biochem Soc Trans 33:1385–1389
Chapter 6
Iron Pathophysiology in Stroke
Abstract Ischemic and hemorrhagic stroke are the common types of stroke that lead
to brain injury neurological deficits and mortality. All forms of stroke remain a seri-
ous health issue, and there is little successful development of drugs for treating stroke.
Incomplete understanding of stroke pathophysiology is considered the main barrier
that limits this research progress. Besides mitochondria and free radical-producing
enzymes, labile iron is an important contributor to oxidative stress. Although iron
regulation and metabolism in cerebral stroke are not fully understood, much progress
has been achieved in recent years. For example, hepcidin has recently been recog-
nized as the principal regulator of systemic iron homeostasis and a bridge between
inflammation and iron regulation. This review discusses recent research progress
in iron pathophysiology following cerebral stroke, focusing molecular regulation of
iron metabolism and potential treatment targets.
6.1 Introduction
Stroke is one of the leading causes of death and long-term severe disability world-
wide. Stroke can be classified into ischemic or hemorrhagic forms according to
its pathophysiological nature. The majority of all stroke cases (around 90%) are
ischemic, which occur due to a sudden decrease or loss of blood supply to the brain
Iron is essential for life as it is needed for many biological processes. In the brain, iron
plays an important role in neurotransmission, myelination, cell division, and oxygen
transport processes [110]. Brain cells maintain iron homeostasis by regulating iron
import, storage, metabolism, and export [50, 51]. Many studies have demonstrated the
role of transferrin, divalent metal transporter 1 (DMT1), ceruloplasmin (CP), ferritin,
and ferroportin1 (FPN1) in the regulation of brain iron [70, 87]. Under physiological
conditions, iron binds to transport proteins such as transferrin. Transferrin-bound iron
is transported into brain cells via receptor-mediated endocytosis. Emerging evidence
suggests that uptake of transferrin-bound iron into the brain cells by transferrin
receptor-mediated endocytosis is the major known pathway of brain iron transport
across the blood–brain barrier (BBB) [87]. This route of iron transport consists of
several steps, including transferrin-iron binding, endocytosis of transferrin-bound
iron, dissociation of iron from transferrin, and translocation of the iron inside brain
cells. Unbound iron can gain access into brain cells through DMT1, but the majority
of iron in brain cells is bound to storage proteins, such as ferritin. CP promotes iron
release from intracellular stores. FPN1 is the first and solely known vertebrate iron
exporter expressed in the brain. It has been shown that FPN1 mediates iron efflux
from brain cells. Since iron import, storage, and export are tightly regulated, any
alterations in these steps may disturb iron homeostasis in the brain resulting in iron
neurotoxicity [2, 87]. Deficiency in FPN1 activity results in iron retention and further
6 Iron Pathophysiology in Stroke 107
iron toxicity [28]. Increased free iron levels have been observed in ischemic and
hemorrhagic brains; however, the exact mechanism behind it is not fully understood
[16, 94].
Iron plays a critical role in oxygen transport [110]. Under ischemic conditions,
required levels of oxygen in the brain increase, resulting in high demand on iron
transport and metabolism inside preserved brain regions [94]. Ferritin-bound iron is
released in response to changes from ischemic insults, including acidosis and inflam-
matory mediators. Many studies have shown that the binding affinity of transferrin
for iron decreases at low pH levels, resulting in dissociation of iron from transferrin
[64]. Upon dissociation, unbound iron can easily gain access to the extracellular
space where it is transported into neurons, resulting in increased intracellular iron
levels [79, 94]. Recent observations have demonstrated that decreased iron efflux
from neuronal cells contributes to putative alterations in iron homeostasis following
ischemic stroke [62]. Furthermore, growing evidence indicates that iron accumu-
lation in neurons following ischemia increases brain susceptibility to iron-induced
damage [41, 64, 94]. In hemorrhagic stroke condition, many studies showed iron
overload in the brain after intracerebral hemorrhage (ICH) [16, 88]. Red blood cell
release and lysis have been considered the major factors mediating iron-induced
brain damage after ICH [53]. Collagenase, which is commonly used to induce ICH,
results in iron overload [113]. Moreover, iron overload may contribute to brain edema
following ICH [43]. However, the potential mechanism, underlying iron-mediated
neurotoxicity, is unsatisfactorily understood.
Many studies have suggested that the severity of neuronal damage after ischemic
stroke is proportional to the magnitude of brain iron accumulation [94]. For example,
Patt et al. demonstrated that gerbils with low brain iron exhibited fewer neurological
deficits compared to those with high brain iron levels [81]. Consistently, rats with
iron overload exhibited about 60% greater infarct volume than rats with lower iron
load [12]. Iron accumulation in rat brains following ischemia [58, 94] induces lipid
peroxidation and neuronal injury by catalyzing conversion of superoxide and hydro-
gen peroxide into highly reactive toxic hydroxyl radicals [54]. Increased formation
of these free radicals results in oxidative stress and subsequent ischemic neuronal
death [11]. In addition, it has been shown that experimental ICH causes brain injury
via iron-induced oxidation and DNA damage [71, 114]. Results from animal studies
108 M. M. A. Almutairi et al.
6.5.2 Hepcidin
Hepcidin was first discovered in human blood and urine as a bactericidal peptide
synthesized in the liver [46, 55]. Since its discovery, many reports have demon-
strated that hepcidin plays a critical role in systemic regulation of iron metabolism
in mammals including human, rat, mouse, and pig [33, 50, 72]. The gene encoding
hepcidin (HAMP) is expressed in a growing list of tissues, including liver, brain,
lungs, intestines, and heart [15, 35, 38, 68, 109]. Hepcidin is a central regulator of
iron homeostasis, but the exact mechanism of how hepcidin regulates iron acquisi-
6 Iron Pathophysiology in Stroke 109
tion has not been fully characterized. Nicolas et al. unambiguously recognized that
inactivation of HAMP results in severe iron accumulation in the liver and pancreas
[76]. Additionally, transgenic mice overexpressing HAMP exhibit a severe anemia
[77]. Human patients with hepcidin mutations suffer from juvenile hemochromato-
sis [89]. Treating cultured cells expressing an FPN1-GFP with hepcidin results in
translocation of FPN1-GFP from the cell surface into lysosomes where it is degraded
[19]. Moreover, the injection of hepcidin into the cerebral ventricle suppresses FPN1
protein levels in different regions of the brain, such as the cerebral cortex, hippocam-
pus, and striatum [24]. Xiong et al. reported that both brain and serum hepcidin was
up-regulated after ICH. This increased expression of hepcidin reduces intracellu-
lar iron efflux from brain microvessel endothelial cells via regulating FPN1 [117].
Studies have shown that hepcidin binds to FPN1 and induces intracellular iron accu-
mulation via interaction with FPN1 and causes its internalization and degradation
in endolysosomes (Fig. 6.1) [19, 20]. It has been identified that residue Cys326 of
FPN1 is required for hepcidin interaction and mutation of this residue induces FPN1
resistance to hepcidin binding [20]. Fernandes et al. proposed that a thiol subunit
of the Cys326 residue initiates a disulfide bond with a cysteine residue in hepcidin
[17, 32]. These findings indicate that increased iron accumulation and decreased
iron efflux might be caused by the action of hepcidin. In physiological conditions,
intercellular iron is mainly stored in ferritin and FPN1 mediates efflux of un-needed
or excess iron. During many pathological conditions, including stroke, hepcidin is
Fig. 6.1 Mechanism of hepcidin action. Hepcidin increases intracellular iron levels by causing the
degradation of FPN1, a cellular membrane protein that export iron to extracellular space
110 M. M. A. Almutairi et al.
up-regulated and binds to FPN1 and causes its internalization and degradation in
endolysosomes. FPN1 degradation leads to iron accumulation in neurons and fur-
ther results in neuronal death.
tive responses to hypoxia. HIF-1 regulates genes that promote energy metabolism,
angiogenesis, and erythropoiesis, all of which are critical in promoting cell survival
in hypoxia. HIF-1 is a heterodimer, consisting of a constitutively expressed β sub-
unit and a regulated α subunit. HIF-1α is preferentially degraded under normoxic
conditions while under hypoxic conditions, it is stabilized, dimerizes with the β sub-
unit, and subsequently induces adaptive gene expression and promotes cell survival.
Under normoxic conditions, HIF-1α is degraded through the ubiquitin–proteasome
system after recognized by the von Hippel Lindau tumor suppressor (pVHL), a com-
ponent of the E3 ubiquitin ligase. The recognition by pVHL requires the action of a
family of HIF prolyl hydroxylase domain (PHD) that hydroxylates proline residues
on HIF-1α in the presence of oxygen. Ferrous ion is a cofactor of the enzyme PHD.
Increase in free iron promotes HIF-1α degradation through the 26S pathway by
elevating its hydroxylation and thus compromises cellular resistance to ischemic
injury (Fig. 6.3). Conversely, reducing iron levels would be lower PHD activity and
increase HIF-1α stability/accumulation. In fact, Hishikawa et al. have reported that
DFO significantly attenuated basilar artery vasospasm and reduction of brainstem
blood flow in a rat model of hemorrhagic stroke by increase in HIF-1α protein level
and HIF-1 activity [52]. Similarly, in ischemic stroke DFO induced HIF-1 DNA bind-
ing and transcription of erythropoietin in vivo and ischemic tolerance [5, 86]. More
recently, Sorond et al. reported that DFO infusion improved cerebrovascular function
in older individuals, possibly by increased the activity of HIF-1-regulated pathways
[99]. Although it is arguable if the protective effect of DFO can be all attributed to
112 M. M. A. Almutairi et al.
Fig. 6.3 New targets for increasing HIF-1 activity. Iron reduction could be an effective pharma-
cological approach to activate HIF-1 and to enhance cerebrovascular function acute and chronic
ischemic conditions. Hepcidin and FPN1 may provide additional targets for increasing HIF-1 levels
that inflammation may drive iron accumulation in cerebral stroke. However, the
hypothesis that hepcidin acts as a link between inflammation and iron neurotoxicity
requires further investigation.
In the last ten years, our understanding of the pathogenesis of stroke-induced brain
injury has advanced. However, therapeutic options for cerebral stroke are still very
limited. Current approved therapies rely mainly on the use of thrombolytic treatment
in acute ischemic cases [104]. Recombinant tissue plasminogen activator (r-tPA) is
the only thrombolytic drug that has been approved for clinical use to reduce neuro-
logical injury and improve the functional recovery of patients with cerebral ischemia
[91]. The time window for effective treatment with r-tPA is limited to three hours
114 M. M. A. Almutairi et al.
after symptom onset. Because the large majority of patients with ischemic stroke
are not aware of initial symptoms, many do not seek care within the three-hour time
frame. Consequently, due to its short therapeutic window and potential hemorrhagic
risk, the number of patients that benefit from this treatment is limited [104]. In
term of ICH, several reports have shown that majority of ICH-patients who received
anticoagulant and antiplatelet treatments had experienced the incidence of severe
anticoagulant-associated ICH [34]. In addition, emerging evidence demonstrated
that antithrombotic drugs such as warfarin increased the risk of ICH by seven folds
and is associated disability and death [30, 40]. This major challenge increases the
demand for safe and effective therapeutic approaches to improve the outcome of this
pathological condition. This section discusses some prospective therapeutic strate-
gies that can protect the brain from the negative consequences of cerebral stroke.
The negative consequences of stroke that could be beneficial therapeutic targets,
including post-stroke inflammation and brain iron accumulation [94, 123, 124].
Hepcidin, a negative regulator of the sole cellular iron exporter FPN1, plays a crucial
role in intracellular iron accumulation [19, 20]. Therefore, therapeutic approaches
that decrease hepcidin synthesis and/or increase FPN1 activity seem to be potential
candidates to treat patient with iron overload disorders [85]. Recent studies have
showed that hepcidin siRNA treatment induces the FPN1 expression and rescues
iron accumulation in cerebral cortex, hippocampus, and corpus striatum of ischemic
mouse brains [24]. In addition, hepcidin knockout mice ICH models exhibit reduced
brain iron accumulation compared with wild-type mice [117]. These findings sug-
gest that iron overload following ischemia and ICH is negatively correlated with
increased FPN1 levels in hepcidin knockout models. Additionally, alteration of
hepcidin-FPN1 interaction with a thiol-reactive compound fursultiamine prevents
FPN1 internalization and degradation via sequestering the Cys326 residue of FPN1,
a necessary residue for hepcidin binding to FPN1 [85]. Consequently, fursultiamine
reduces intracellular iron accumulation via enhanced cellular iron export despite the
presence of hepcidin [36]. As discussed previously, activation of IL6/STAT3 path-
way induces hepcidin transcription [24]. Therefore, targeting IL6/STAT3 pathway
with anti-IL6 antibodies can down-regulate hepcidin expression. Siltuximab and
tocilizumab, anti-IL6 antibody drugs, are in clinical use to reduce hepcidin effects
and improve cellular iron export in patients with anemia [85]. Recent studies have
116 M. M. A. Almutairi et al.
shown that anti-IL6 antibody significantly decreases the expression of IL-6 protein in
the brain of ischemic animal models [102]. Furthermore, anti-IL6 antibody rescues
the blood–brain barrier from ischemic injury in the fetal sheep model [120]. In addi-
tion, urocortin, anti-inflammatory neuropeptide, suppresses inflammatory cytokine
production (e.g., IL-6 and TNF-α) and reduces neurological deficits following ICH
[63]. Taken together, targeting hepcidin and/or corresponding pathways could be an
effective therapeutic approach to protect the brain from stroke injury.
Inflammation and iron accumulation play a crucial role in neuronal injuries caused by
ischemic stroke. Hepcidin binds FPN1 and reduces its activity in excreting excess iron
from the brain cells. The major factors that positively regulate hepcidin include iron
status and inflammation. Modifying iron metabolism and post-stroke inflammation
can reduce brain injury in stroke and the hepcidin pathway seems to be a promising
molecular target for stroke therapy.
Increased hepcidin expression has emerged as a causative factor in several com-
mon iron disorders such as cerebral stroke. Targeting hepcidin pathways and/or
interfering hepcidin–FPN1 interaction have become promising approaches for the
development of novel therapeutics for iron disorders. However, multiple potential
opportunities for cross communication among these pathways can cause off-target
effects such as an increased risk of infection. For example, IL-6 receptor-targeted ther-
apeutics impair host defense. Moreover, interfering hepcidin–FPN1 interaction by
fursultiamine antagonizes hepcidin effects rather than reducing its expression. There-
fore, lowering hepcidin expression directly in cerebral stroke where it is elevated may
have less possible off-target effects in comparison with other therapeutic approaches
mentioned previously. In addition, the direct targeting of hepcidin in stroke models
would help us to understand the specific role of hepcidin in stroke-induced brain
injury. The major challenge of systemic administration of anti-hepcidin is that some
negative effects need to be taken into account. Deficiency of systemic hepcidin results
in increased serum iron, dietary iron hyperabsorption, and iron-dependent oxidative
stress. To overcome this challenge, anti-hepcidin could be delivered into the stroke-
injured brain by using microinjection or nanoparticles as drug delivery systems. In
the following, some experimental approaches are proposed in order to refine our
understanding of the mechanistic role of hepcidin in cerebral stroke.
In summary, these future directions may improve our understating of the roles
of hepcidin in iron-mediated neurotoxicity following cerebral stroke. Based on the
results, novel therapeutic interventions might be developed to protect the brain from
stroke injury.
References
21. Deb P, Sharma S, Hassan KM (2010) Pathophysiologic mechanisms of acute ischemic stroke:
an overview with emphasis on therapeutic significance beyond thrombolysis. Pathophysiology
17:197–218
22. Deiner S, Silverstein JH (2009) Postoperative delirium and cognitive dysfunction. Br J Anaesth
103(Suppl 1):i41–i46
23. Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV (2007) Absence of the chemokine
receptor CCR23 protects against cerebral ischemia/reperfusion injury in mice. Stroke
38:1345–1353
24. Ding H, Yan CZ, Shi H, Zhao YS, Chang SY, Yu P, Wu WS, Zhao CY, Chang YZ, Duan XL
(2011) Hepcidin is involved in iron regulation in the ischemic brain. PLoS ONE 6:e25324
25. Dirnagl U, Iadecola C, Moskowitz MA (1999) Pathobiology of ischaemic stroke: an integrated
view. Trends Neurosci 22:391–397
26. Donnan GA, Fisher M, Macleod M, Davis SM (2008) Stroke. Lancet 371:1612–1623
27. Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt SJ, Moynihan J, Paw BH, Drejer A, Barut
B, Zapata A, Law TC, Brugnara C, Lux SE, Pinkus GS, Pinkus JL, Kingsley PD, Palis
J, Fleming MD, Andrews NC, Zon LI (2000) Positional cloning of zebrafish ferroportin1
identifies a conserved vertebrate iron exporter. Nature 403:776–781
28. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S, Andrews NC (2005) The
iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab 1:191–200
29. Ekdahl CT, Kokaia Z, Lindvall O (2009) Brain inflammation and adult neurogenesis: the dual
role of microglia. Neuroscience 158:1021–1029
30. Fang MC, Go AS, Chang Y, Hylek EM, Henault LE, Jensvold NG, Singer DE (2007) Death
and disability from warfarin-associated intracranial and extracranial hemorrhages. Am J Med
120:700–705
31. Feng Y, Leblanc MH, Leblanc EB, Parker CC, Fratkin JD, Qian XB, Patel DM, Huang M,
Smith EE, Vig PJ (2000) Desmethyl tirilazad improves neurologic function after hypoxic
ischemic brain injury in piglets. Crit Care Med 28:1431–1438
32. Fernandes A, Preza GC, Phung Y, de Domenico I, Kaplan J, Ganz T, Nemeth E (2009) The
molecular basis of hepcidin-resistant hereditary hemochromatosis. Blood 114:437–443
33. Fleming RE, Sly WS (2002) Mechanisms of iron accumulation in hereditary hemochromato-
sis. Annu Rev Physiol 64:663–680
34. Fogarty Mack P (2014) Intracranial haemorrhage: therapeutic interventions and anaesthetic
management. Br J Anaesth 113(Suppl 2):ii17–ii25
35. Fujita N, Sugimoto R, Takeo M, Urawa N, Mifuji R, Tanaka H, Kobayashi Y, Iwasa M,
Watanabe S, Adachi Y, Kaito M (2007) Hepcidin expression in the liver: relatively low level
in patients with chronic hepatitis C. Mol Med 13:97–104
36. Fung E, Sugianto P, Hsu J, Damoiseaux R, Ganz T, Nemeth E (2013) High-throughput screen-
ing of small molecules identifies hepcidin antagonists. Mol Pharmacol 83:681–690
37. Ganz T (2005) Cellular iron: ferroportin is the only way out. Cell Metab 1:155–157
38. Ganz T (2005) Hepcidin–a regulator of intestinal iron absorption and iron recycling by
macrophages. Best Pract Res Clin Haematol 18:171–182
39. Ganz T, Nemeth E (2012) Hepcidin and iron homeostasis. Biochim Biophys Acta
1823:1434–1443
40. Garcia-Rodriguez LA, Gaist D, Morton J, Cookson C, Gonzalez-Perez A (2013) Antithrom-
botic drugs and risk of hemorrhagic stroke in the general population. Neurology 81:566–574
41. Garcia-Yebenes I, Sobrado M, Moraga A, Zarruk JG, Romera VG, Pradillo JM, de la Ossa NP,
Moro MA, Davalos A, Lizasoain I (2012) Iron overload, measured as serum ferritin, increases
brain damage induced by focal ischemia and early reperfusion. Neurochem Int 61:1364–1369
42. Garcia JM, Stillings SA, Leclerc JL, Phillips H, Edwards NJ, Robicsek SA, Hoh BL, Blackburn
S, Dore S (2017) Role of interleukin-10 in acute brain injuries. Front Neurol 8:244
43. Garton T, Keep RF, Hua Y, Xi G (2016) Brain iron overload following intracranial haemor-
rhage. Stroke Vasc Neurol 1:172–184
6 Iron Pathophysiology in Stroke 119
44. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox
CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD,
Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH,
Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER 3rd, Moy CS,
Mussolino ME, Neumar RW, Nichol G, Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein
J, Towfighi A, Turan TN, Virani SS, Wong ND, Woo D, Turner MB (2014) Heart disease
and stroke statistics–2014 update: a report from the American Heart Association. Circulation
129:e28–e292
45. Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY, Wang ZY (2013) Deferoxamine
inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse
brain. Neurochem Int 62:165–172
46. Guo P, Cui R, Chang YZ, Wu WS, Qian ZM, Yoshida K, Qiao YT, Takeda S, Duan XL
(2009) Hepcidin, an antimicrobial peptide is downregulated in ceruloplasmin-deficient mice.
Peptides 30:262–266
47. Hall ED (1992) Novel inhibitors of iron-dependent lipid peroxidation for neurodegenerative
disorders. Ann Neurol 32(Suppl):S137–S142
48. Hanson LR, Roeytenberg A, Martinez PM, Coppes VG, Sweet DC, Rao RJ, Marti DL, Hoek-
man JD, Matthews RB, Frey WH 2nd, Panter SS (2009) Intranasal deferoxamine provides
increased brain exposure and significant protection in rat ischemic stroke. J Pharmacol Exp
Ther 330:679–686
49. Hemorrhagic Stroke Academia Industry Roundtable Participants (2018) Unmet needs and
challenges in clinical research of intracerebral hemorrhage. Stroke 49:1299–1307
50. Hentze MW, Muckenthaler MU, Andrews NC (2004) Balancing acts: molecular control of
mammalian iron metabolism. Cell 117:285–297
51. Hentze MW, Muckenthaler MU, Galy B, Camaschella C (2010) Two to tango: regulation of
Mammalian iron metabolism. Cell 142:24–38
52. Hishikawa T, Ono S, Ogawa T, Tokunaga K, Sugiu K, Date I (2008) Effects of deferoxamine-
activated hypoxia-inducible factor-1 on the brainstem after subarachnoid hemorrhage in rats.
Neurosurgery 62:232–240 (discussion 240-1)
53. Hua Y, Keep RF, Hoff JT, Xi G (2007) Brain injury after intracerebral hemorrhage: the role
of thrombin and iron. Stroke 38:759–762
54. Jomova K, Valko M (2011) Advances in metal-induced oxidative stress and human disease.
Toxicology 283:65–87
55. Kanda J, Mizumoto C, Kawabata H, Tsuchida H, Tomosugi N, Matsuo K, Uchiyama T (2008)
Serum hepcidin level and erythropoietic activity after hematopoietic stem cell transplantation.
Haematologica 93:1550–1554
56. Kidwell CS, Saver JL, Starkman S, Duckwiler G, Jahan R, Vespa P, Villablanca JP, Liebeskind
DS, Gobin YP, Vinuela F, Alger JR (2002) Late secondary ischemic injury in patients receiving
intraarterial thrombolysis. Ann Neurol 52:698–703
57. Kim JS, Gautam SC, Chopp M, Zaloga C, Jones ML, Ward PA, Welch KM (1995) Expression
of monocyte chemoattractant protein-1 and macrophage inflammatory protein-1 after focal
cerebral ischemia in the rat. J Neuroimmunol 56:127–134
58. Kondo Y, Ogawa N, Asanuma M, Ota Z, Mori A (1995) Regional differences in late-onset
iron deposition, ferritin, transferrin, astrocyte proliferation, and microglial activation after
transient forebrain ischemia in rat brain. J Cereb Blood Flow Metab 15:216–226
59. Lakhan SE, Kirchgessner A, Hofer M (2009) Inflammatory mechanisms in ischemic stroke:
therapeutic approaches. J Transl Med 7:97
60. Lampl Y, Boaz M, Gilad R, Lorberboym M, Dabby R, Rapoport A, Anca-Hershkowitz M,
Sadeh M (2007) Minocycline treatment in acute stroke: an open-label, evaluator-blinded study.
Neurology 69:1404–1410
61. Lee P, Peng H, Gelbart T, Wang L, Beutler E (2005) Regulation of hepcidin transcription by
interleukin-1 and interleukin-6. Proc Natl Acad Sci U S A 102:1906–1910
120 M. M. A. Almutairi et al.
62. Li L, Li YW, Zhao JY, Liu YZ, Holscher C (2009) Quantitative analysis of iron concentration
and expression of ferroportin 1 in the cortex and hippocampus of rats induced by cerebral
ischemia. J Clin Neurosci 16:1466–1472
63. Liew HK, Pang CY, Hsu CW, Wang MJ, Li TY, Peng HF, Kuo JS, Wang JY (2012) Systemic
administration of urocortin after intracerebral hemorrhage reduces neurological deficits and
neuroinflammation in rats. J Neuroinflammation 9:13
64. Lipscomb DC, Gorman LG, Traystman RJ, Hurn PD (1998) Low molecular weight iron in
cerebral ischemic acidosis in vivo. Stroke 29:487–492 (discussion 493)
65. Lucas SM, Rothwell NJ, Gibson RM (2006) The role of inflammation in CNS injury and
disease. Br J Pharmacol 147(Suppl 1):S232–S240
66. Mason SE, Noel-Storr A, Ritchie CW (2010) The impact of general and regional anesthe-
sia on the incidence of post-operative cognitive dysfunction and post-operative delirium: a
systematic review with meta-analysis. J Alzheimers Dis 22(Suppl 3):67–79
67. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Barrow D, Miret S, Bomford A, Peters TJ,
Farzaneh F, Hediger MA, Hentze MW, Simpson RJ (2000) A novel duodenal iron-regulated
transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Mol Cell
5:299–309
68. Merle U, Fein E, Gehrke SG, Stremmel W, Kulaksiz H (2007) The iron regulatory peptide
hepcidin is expressed in the heart and regulated by hypoxia and inflammation. Endocrinology
148:2663–2668
69. Monk TG, Weldon BC, Garvan CW, Dede DE, van der Aa MT, Heilman KM, Gravenstein JS
(2008) Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology
108:18–30
70. Moos T, Rosengren Nielsen T, Skjorringe T, Morgan EH (2007) Iron trafficking inside the
brain. J Neurochem 103:1730–1740
71. Nakamura T, Keep RF, Hua Y, Nagao S, Hoff JT, Xi G (2006) Iron-induced oxidative brain
injury after experimental intracerebral hemorrhage. Acta Neurochir Suppl 96:194–198
72. Nemeth E, Ganz T (2006) Regulation of iron metabolism by hepcidin. Annu Rev Nutr
26:323–342
73. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, Ganz T (2004) IL-
6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory
hormone hepcidin. J Clin Invest 113:1271–1276
74. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan
J (2004) Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its
internalization. Science 306:2090–2093
75. Ni W, Okauchi M, Hatakeyama T, Gu Y, Keep RF, Xi G, Hua Y (2015) Deferoxamine reduces
intracerebral hemorrhage-induced white matter damage in aged rats. Exp Neurol 272:128–134
76. Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, Vaulont S (2001)
Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory
factor 2 (USF2) knockout mice. Proc Natl Acad Sci U S A 98:8780–8785
77. Nicolas G, Viatte L, Lou DQ, Bennoun M, Beaumont C, Kahn A, Andrews NC, Vaulont
S (2003) Constitutive hepcidin expression prevents iron overload in a mouse model of
hemochromatosis. Nat Genet 34:97–101
78. Ooboshi H, Ibayashi S, Shichita T, Kumai Y, Takada J, Ago T, Arakawa S, Sugimori H,
Kamouchi M, Kitazono T, Iida M (2005) Postischemic gene transfer of interleukin-10 protects
against both focal and global brain ischemia. Circulation 111:913–919
79. Palmer C, Menzies SL, Roberts RL, Pavlick G, Connor JR (1999) Changes in iron histochem-
istry after hypoxic-ischemic brain injury in the neonatal rat. J Neurosci Res 56:60–71
80. Palmer C, Roberts RL, Bero C (1994) Deferoxamine posttreatment reduces ischemic brain
injury in neonatal rats. Stroke 25:1039–1045
81. Patt A, Horesh IR, Berger EM, Harken AH, Repine JE (1990) Iron depletion or chelation
reduces ischemia/reperfusion-induced edema in gerbil brains. J Pediatr Surg 25:224–227
(discussion 227-8)
6 Iron Pathophysiology in Stroke 121
82. Perna R, Temple J (2015) Rehabilitation outcomes: ischemic versus hemorrhagic strokes.
Behav Neurol 2015:891651
83. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, Loreal O (2001) A new mouse
liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin,
is overexpressed during iron overload. J Biol Chem 276:7811–7819
84. Poggiali E, Cassinerio E, Zanaboni L, Cappellini MD (2012) An update on iron chelation
therapy. Blood Transfus 10:411–422
85. Poli M, Asperti M, Ruzzenenti P, Regoni M, Arosio P (2014) Hepcidin antagonists for potential
treatments of disorders with hepcidin excess. Front Pharmacol 5:86
86. Prass K, Ruscher K, Karsch M, Isaev N, Megow D, Priller J, Scharff A, Dirnagl U, Meisel
A (2002) Desferrioxamine induces delayed tolerance against cerebral ischemia in vivo and
in vitro. J Cereb Blood Flow Metab 22:520–525
87. Qian ZM, Shen X (2001) Brain iron transport and neurodegeneration. Trends Mol Med
7:103–108
88. Righy C, Turon R, Freitas G, Japiassu AM, Faria Neto HCC, Bozza M, Oliveira MF, Bozza FA
(2018) Hemoglobin metabolism by-products are associated with an inflammatory response
in patients with hemorrhagic stroke. Rev Bras Ter Intensiva 30:21–27
89. Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D, Christakis J, Loukopoulos D, Cam-
aschella C (2003) Mutant antimicrobial peptide hepcidin is associated with severe juvenile
hemochromatosis. Nat Genet 33:21–22
90. Rosenthal RE, Chanderbhan R, Marshall G, Fiskum G (1992) Prevention of post-ischemic
brain lipid conjugated diene production and neurological injury by hydroxyethyl starch-
conjugated deferoxamine. Free Radic Biol Med 12:29–33
91. Roth JM (2011) Recombinant tissue plasminogen activator for the treatment of acute ischemic
stroke. Proc (Bayl Univ Med Cent) 24:257–259
92. Saleppico S, Mazzolla R, Boelaert JR, Puliti M, Barluzzi R, Bistoni F, Blasi E (1996) Iron reg-
ulates microglial cell-mediated secretory and effector functions. Cell Immunol 170:251–259
93. Selim M (2010) Treatment with the iron chelator, deferoxamine mesylate, alters serum markers
of oxidative stress in stroke patients. Transl Stroke Res 1:35–39
94. Selim MH, Ratan RR (2004) The role of iron neurotoxicity in ischemic stroke. Ageing Res
Rev 3:345–353
95. Seymour DG, Severn AM (2009) Cognitive dysfunction after surgery and anaesthesia: what
can we tell the grandparents? Age Ageing 38:147–150
96. Shichita T, Ito M, Yoshimura A (2014) Post-ischemic inflammation regulates neural damage
and protection. Front Cell Neurosci 8:319
97. Soloniuk DS, Perkins E, Wilson JR (1992) Use of allopurinol and deferoxamine in cellular
protection during ischemia. Surg Neurol 38:110–113
98. Soriano SG, Amaravadi LS, Wang YF, Zhou H, Yu GX, Tonra JR, Fairchild-Huntress V, Fang
Q, Dunmore JH, Huszar D, Pan Y (2002) Mice deficient in fractalkine are less susceptible to
cerebral ischemia-reperfusion injury. J Neuroimmunol 125:59–65
99. Sorond FA, Tan CO, Larose S, Monk AD, Fichorova R, Ryan S, Lipsitz LA (2015) Deferoxam-
ine, cerebrovascular hemodynamics, and vascular aging: potential role for hypoxia-inducible
transcription factor-1-regulated pathways. Stroke 46:2576–2583
100. Steinmetz J, Christensen KB, Lund T, Lohse N, Rasmussen LS, Group I (2009) Long-term
consequences of postoperative cognitive dysfunction. Anesthesiology 110:548–555
101. Tan G, Liu L, He Z, Sun J, Xing W, Sun X (2016) Role of hepcidin and its downstream
proteins in early brain injury after experimental subarachnoid hemorrhage in rats. Mol Cell
Biochem 418:31–38
102. Tang YH, Vital S, Russell J, Seifert H, Neil Granger D (2015) Interleukin-6 mediates enhanced
thrombus development in cerebral arterioles following a brief period of focal brain ischemia.
Exp Neurol 271:351–357
103. Taylor RA, Chang CF, Goods BA, Hammond MD, Mac Grory B, Ai Y, Steinschneider AF,
Renfroe SC, Askenase MH, McCullough LD, Kasner SE, Mullen MT, Hafler DA, Love JC,
Sansing LH (2017) TGF-beta1 modulates microglial phenotype and promotes recovery after
intracerebral hemorrhage. J Clin Invest 127:280–292
122 M. M. A. Almutairi et al.
104. Tobin MK, Bonds JA, Minshall RD, Pelligrino DA, Testai FD, Lazarov O (2014) Neurogenesis
and inflammation after ischemic stroke: what is known and where we go from here. J Cereb
Blood Flow Metab 34:1573–1584
105. Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP, Arredondo M, Gonzalez-Billault C, Nunez
MT (2013) Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes
iron accumulation in central nervous system cells. J Neurochem 126:541–549
106. Verga Falzacappa MV, Vujicspasic M, Kessler R, Stolte J, Hentze MW, Muckenthaler MU
(2007) STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation. Blood
109:353–358
107. Wang G, Hu W, Tang Q, Wang L, Sun XG, Chen Y, Yin Y, Xue F, Sun Z (2016) Effect
comparison of both iron chelators on outcomes, iron deposit, and iron transporters after
intracerebral hemorrhage in rats. Mol Neurobiol 53:3576–3585
108. Wang Q, Du F, Qian ZM, Ge XH, Zhu L, Yung WH, Yang L, Ke Y (2008) Lipopolysaccharide
induces a significant increase in expression of iron regulatory hormone hepcidin in the cortex
and substantia nigra in rat brain. Endocrinology 149:3920–3925
109. Wang SM, Fu LJ, Duan XL, Crooks DR, Yu P, Qian ZM, Di XJ, Li J, Rouault TA, Chang YZ
(2010) Role of hepcidin in murine brain iron metabolism. Cell Mol Life Sci 67:123–133
110. Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L (2014) The role of iron in brain ageing
and neurodegenerative disorders. Lancet Neurol 13:1045–1060
111. Wilkinson DA, Pandey AS, Thompson BG, Keep RF, Hua Y, Xi G (2018) Injury mechanisms
in acute intracerebral hemorrhage. Neuropharmacology 134:240–248
112. Wrighting DM, Andrews NC (2006) Interleukin-6 induces hepcidin expression through
STAT3. Blood 108:3204–3209
113. Wu H, Wu T, Xu X, Wang J, Wang J (2011) Iron toxicity in mice with collagenase-induced
intracerebral hemorrhage. J Cereb Blood Flow Metab 31:1243–1250
114. Wu J, Hua Y, Keep RF, Schallert T, Hoff JT, Xi G (2002) Oxidative brain injury from
extravasated erythrocytes after intracerebral hemorrhage. Brain Res 953:45–52
115. Xiao L, Li XQ, Zhang HX (2004) Effects of naoyian granule on the expression of IL-6 mRNA
and protein in the experimental intracerebral hemorrhagic brain in rats. Zhong Nan Da Xue
Xue Bao Yi Xue Ban 29:177–180
116. Xie J, Brayne C, Matthews FE, Medical Research Council Cognitive F, Ageing Study C
(2008) Survival times in people with dementia: analysis from population based cohort study
with 14 year follow-up. BMJ 336:258–262
117. Xiong XY, Liu L, Wang FX, Yang YR, Hao JW, Wang PF, Zhong Q, Zhou K, Xiong A,
Zhu WY, Zhao T, Meng ZY, Wang YC, Gong QW, Liao MF, Wang J, Yang QW (2016)
Toll-like receptor 4/MyD88-mediated signaling of hepcidin expression causing brain iron
accumulation, oxidative injury, and cognitive impairment after intracerebral hemorrhage.
Circulation 134:1025–1038
118. Yamasaki Y, Matsuura N, Shozuhara H, Onodera H, Itoyama Y, Kogure K (1995) Interleukin-
1 as a pathogenetic mediator of ischemic brain damage in rats. Stroke 26:676–680 (discussion
681)
119. Yang L, Zhang B, Yin L, Cai B, Shan H, Zhang L, Lu Y, Bi Z (2011) Tanshinone IIA prevented
brain iron dyshomeostasis in cerebral ischemic rats. Cell Physiol Biochem 27:23–30
120. Zhang J, Sadowska GB, Chen X, Park SY, Kim JE, Bodge CA, Cummings E, Lim YP, Makeyev
O, Besio WG, Gaitanis J, Banks WA, Stonestreet BS (2015) Anti-IL-6 neutralizing antibody
modulates blood-brain barrier function in the ovine fetus. FASEB J 29:1739–1753
121. Zhang XY, Cao JB, Zhang LM, Li YF, Mi WD (2015) Deferoxamine attenuates
lipopolysaccharide-induced neuroinflammation and memory impairment in mice. J Neuroin-
flammation 12:20
122. Zhao Y, Rempe DA (2011) Prophylactic neuroprotection against stroke: low-dose, prolonged
treatment with deferoxamine or deferasirox establishes prolonged neuroprotection indepen-
dent of HIF-1 function. J Cereb Blood Flow Metab 31:1412–1423
123. Zheng Z, Yenari MA (2004) Post-ischemic inflammation: molecular mechanisms and thera-
peutic implications. Neurol Res 26:884–892
6 Iron Pathophysiology in Stroke 123
124. Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW (2014) Inflammation in intracerebral
hemorrhage: from mechanisms to clinical translation. Prog Neurobiol 115:25–44
125. Zohn IE, de Domenico I, Pollock A, Ward DM, Goodman JF, Liang X, Sanchez AJ, Niswander
L, Kaplan J (2007) The flatiron mutation in mouse ferroportin acts as a dominant negative to
cause ferroportin disease. Blood 109:4174–4180
Chapter 7
Iron Pathophysiology in Friedreich’s
Ataxia
Kuanyu Li
Abstract Friedreich’s ataxia (FRDA) is a degenerative disease that affects both the
central and the peripheral nervous systems and non-neural tissues including, mainly,
heart, and endocrine pancreas. It is an autosomal recessive disease caused by a
GAA triplet-repeat localized within an Alu sequence element in intron 1 of frataxin
(FXN) gene, which encodes a mitochondrial protein FXN. This protein is essential
for mitochondrial function by the involvement of iron–sulfur cluster biogenesis. The
effects of its deficiency also include disruption of cellular, particularly mitochondrial,
iron homeostasis, i.e., relatively more iron accumulated in mitochondria and less
iron presented in cytosol. Though iron toxicity is commonly thought to be mediated
via Fenton reaction, oxidative stress seems not to be the main problem to result
in detrimental effects on cell survival, particularly neuron survival. Therefore, the
basic research on FXN function is urgently demanded to understand the disease.
This chapter focuses on the outcome of FXN expression, regulation, and function
in cellular or animal models of FRDA and on iron pathophysiology in the affected
tissues. Finally, therapeutic strategies based on the control of iron toxicity and iron
cellular redistribution are considered. The combination of multiple therapeutic targets
including iron, oxidative stress, mitochondrial function, and FXN regulation is also
proposed.
K. Li (B)
Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing
210093, People’s Republic of China
e-mail: [email protected]
unaided. The disease occurs mainly in Caucasians. It is believed that the restricted
distribution in Caucasians is due to a single founder mutation that occurred between
500 and 800 generations ago [1].
The essential clinical and pathological features of the disease include the degen-
eration in the spinal cord of the posterior columns, which contain axons of dorsal-
root-ganglia sensory neurons. Most patients succumb to cardiomyopathy, and many
become diabetic during the course of the disease. Typically, cognitive functions are
preserved well. The presence of comorbid depression in FRDA is recently recog-
nized [2]. In 1996, came the breakthrough identification of the disease gene, frataxin
(FXN), for FRDA [3].
The last 20 years have seen remarkable progress in elucidating the function of the
encoded protein, FXN, which localizes to the mitochondria [4] and is involved in
iron-sulfur cluster biogenesis [5]. The focus of this chapter is on iron dysregulation
in FRDA, with a particular emphasis on the most recent findings.
Ages of onset and death as a function of GAA trinucleotide repeat expansions are
closely correlated with expression levels of FXN. This GAA expansion ranges from
fewer than 200 to as many as 1500 repeats, in which size of the expansion correlates
negatively with expression of the encoded protein and positively with the ages of onset
and severity of the disease. This expansion leads to transcriptional silencing of FXN
through a commonly accepted mechanism involving modifications of the chromatin
structure of the locus, resulting in expression of a structurally and functionally normal
FXN but at levels that are estimated at 5–30% of normal [8, 9]. The decreased histone
acetylation and extensive methylation of CpG regions upstream of the GAA repeats
are observed in patient cell lines and tissues [10, 11], suggesting that the enhanced
heterochromatin formation might impede the transcription of FXN, leading to the
lower levels of FXN protein [12–14]. Studies employing an experimental histone
7 Iron Pathophysiology in Friedreich’s Ataxia 127
Studies suggested that FXN might be the iron donor or iron chaperone for the assem-
bly of the Fe–S cluster [25, 34–36]. It was proposed in early studies that FXN could
act as a metal chaperone because the structure indicated the iron-binding sites of
FXN [37] and the purified protein could coordinate iron [38]. There is also uncer-
tainty on the number of metal ions that are bound per FXN monomer, whereas 2
ferrous iron or 6–7 ferric iron ions per FXN monomer were suggested using a group
of exposed acidic residues [37, 38]. These amino acids are different from the canon-
ical iron-binding motifs, which normally are cysteine and/or histidine. The rational
for the iron-binding activity of FXN was proposed by the interaction between FXN
and ISCU [30] and between FXN and ferrochelatase (FECH) [39]. The former com-
plex facilitates the transfer of iron from holo-FXN to nucleation sites for [2Fe–2S]
cluster formation on ISU [38], while the latter complex mediates iron insertion into
protoporphyrin IX to make heme [40]. The destination of iron delivery by FXN
was determined by the cellular physiological activities, i.e., mainly to FECH for
heme biosynthesis in erythrocytes by down-regulation of FXN and to both ISCU and
FECH in normal cells to fulfill the two major iron-dependent pathways [40]. How-
ever, a recent study demonstrated that no changes in heme synthesis was observed in
human FRDA erythroid progenitor cells compared with healthy control cells, which
was supported by that FRDA patients do not have abnormal hematological symp-
toms. As for the distinct high binding affinities of holo-FXN to the target protein
FECH, the binding between might be a negative mechanism for removal of FXN
during erythropoiesis. In non-erythrocytes, it is relevant that holo-FXN delivers iron
to ISCU, particularly in cells with highly expressed FXN, for instance in FRDA-
affected tissues.
More recently, Isaya and colleagues found that iron binding to in vitro purified
FXN promoted its oligomerization to form a complex of high molecular weight
(850–1100 kDa) [42, 43]. These oligomeric forms resemble ferritin structure con-
7 Iron Pathophysiology in Friedreich’s Ataxia 129
taining thousands of iron ions for iron storage [42, 44]. Therefore, FXN is proposed
to act as ferritin for mitochondrial iron storage. Indeed, the ferroxidase activity of
FXN was revealed in yeast [45, 46], further supporting the iron-binding activity.
However, another study revealed that human mitochondrial ferritin improves respi-
ratory function in yeast mutants deficient in iron–sulfur cluster biogenesis, but is
not a functional homologue of yeast FXN [47]. Concerning the iron-binding activity
and iron-induced oligomerization, more detailed investigation has elucidated that a
[FXN42–210]24 :[NFS1]24 :[ISD11]24 :[ISCU]24 complex is consistent with the mea-
sured 1:1:1:1 stoichiometry of its four components [35]. This structure of the complex
does not only recapitulate the known mechanism for sulfur donation from NFS1 to
ISCU but also provides a path for iron donation from FXN42–210 to ISCU. How-
ever, isoforms FXN42–210 and FXN81–210 have strikingly different biochemical
properties (reviewed in [48]). The former one has not yet accepted abundant in heart,
cerebellum, and dividing lymphoblasts, and the later one only forms monomeric
complex in vivo. Overall, it was suggested that the iron binding of the mature form
is served for delivery of iron to biochemical pathways, while FXN42–210 could be
able to oligomerize and store iron as ferritin.
The role of FXN in orchestrating Fe–S cluster formation is widely accepted. Iron is
considered first to be imported into the mitochondria by mitoferrins (MFRN1 and
MFRN2) [49]. MFRN1 is inducibly expressed during erythropoiesis, and MFRN2 is
ubiquitously expressed in all tissues, probably, for basal mitochondrial iron uptake.
The iron is then majorly used for Fe–S cluster and heme biogenesis within mito-
chondria though how the iron was delivered into the process is in debate. De novo
Fe–S cluster biogenesis involves assembly of inorganic sulfur and iron on a scaffold
protein ISCU, with which a complex consists of NFS1-ISD11, the cysteine desul-
furase providing the sulfur, and FXN. A few studies support that FXN regulates the
NFS1 activity and the entry of iron within the complex via stabilizing the complex
[50–52], which is in consistence with the observation that the binding of FXN with
NFS1 is not iron dependent [23]. These results would not support FXN functions
as an iron donor for Fe–S cluster biosynthesis, rather than as an allosteric regulator
for enhancing NFS1 function to form persulfide on NFS1 and to transfer sulfur from
NFS1 to ISCU as an initial step in Fe–S cluster biosynthesis [52]. Still, how FXN
stabilizes the quaternary complex of core components and controls iron entry to the
complex and how iron gets to the entry and is transferred to scaffold protein ISCU
are mysterious.
Though the functions of FXN have been proposed many, for instance, involvements
against oxidative stress and heme biogenesis, they may be the secondary effects of
130 K. Li
Fe–S cluster deficiency. The lack of FXN prevents the assembly of Fe–S clusters with
consecutive mitochondrial iron overload [53]. The toxic radicals through the Fenton
reaction are generated at an excessively high level in FXN-deficient mitochondria.
Meanwhile, due to the lack of Fe–S clusters as important cofactors in complex I, II,
and III of electron transport chain, the respiratory chain is defective, which makes
reactive oxygen species (ROS) produced more severely. These ROS can oxidize
ferrous iron and disrupt the cellular redox homeostasis. The consequence will be the
damage of macromolecules including DNA, proteins, and lipids, which may be the
pathological mechanism of FRDA. Interestingly, not all mitochondria-rich tissues
are affected in FRDA patients, but clearly cerebellum, spinal cord, and heart.
Because ferrochelatase, the last enzyme for heme biogenesis, needs Fe–S cluster to
gain the enzymatic activity for insertion of iron into protoporphyrin IX to form heme,
the lack of Fe–S cluster induced by FXN deficiency would affect heme biogenesis.
Therefore, FRDA patients would develop hematological disorder. FXN was found to
interact with ferrochelatase, which hints that FXN delivers iron to ferrochelatase to
produce heme in cells, particularly in erythroblasts. However, FRDA patients do not
present typical hematological disorder [41], which suggests that FXN may also play
a role in determining the recipient of this prosthetic group after assembly, favorably
being transferred to mitochondrial respiratory complexes.
The functions of FXN have been proposed involving biogenesis of iron–sulfur clus-
ters, iron chaperoning, iron storage, and control of iron-mediated oxidative tissue
damage (reviewed in [54]). As demonstrated in knockout animals, complete absence
of FXN leads to early embryonic lethality [55], indicating that at least some FXN
function is essential for survival. Despite the difficulty in generating perfect FRDA
models, a multitude of complementary models have been generated enabling signif-
icant advances in understanding the function of FXN in model organisms as yeast,
fruit fly, nematode, and mouse.
In yeast, the Yfh1 protein (encoded by yeast FXN homolog Yfh1p) is confirmed
to be involved in iron homeostasis and mitochondrial function [56]. The deletion of
the Yfh1 gene makes mitochondrial iron accumulation (up to 10 times more than the
wild strain) makes the cells highly sensitive to oxidative stress, and also makes the
strain unable to grow on a non-fermentable source of carbon [53, 56], suggesting that
it could not carry out oxidative phosphorylation. In drosophila, FXN homolog (dfh)
was identified, a much smaller genomic region than the human gene [57]. Its genomic
composition just includes a single intron dividing the coding region into two exons.
Moderate systemic reduction of dfh expression with RNA interference (RNAi) to
levels of 30% of normal dfh-mRNA represents the situation in patients of FRDA, in
which flies develop with normal embryonic stage, but shortened mean and maximum
life span of 60 and 32%, respectively [58]. Reduction of FXN drastically compro-
mises aconitase activity and increases the levels of reactive oxygen species (ROS)
7 Iron Pathophysiology in Friedreich’s Ataxia 131
Iron relation was first evidenced for myocardial iron deposits in FRDA hearts [72].
Much later, iron involvement was recognized by the finding of deficiencies of aconi-
tase and succinate dehydrogenase [28], both of which are mitochondrial iron–sulfur
cluster-containing enzymes.
FXN deficiency leads to mitochondrial iron overload, defective energy supply,
and generation of reactive oxygen species [67] (also see reviews [34, 73]). To date,
no data provide a clear answer on the nature of the iron that is accumulated in affected
mitochondria, but few interesting hints are available. The deposited mitochondrial
iron was identified to be a biomineral aggregate distinctly different from mammalian
ferritin [74] due to low expression of mitochondrial ferritin and high expression of
mitochondrial importer mitoferrin 2 (MFRN2) [75]. What drives high expression of
mitoferrin and how the excess toxic iron is trapped in the mitochondrial matrix are
still unknown. It seems that mitochondrial iron overload is commonly taken place
when Fe–S cluster biogenesis or transfer is disturbed. A similar increase of MFRN2
mRNA has been revealed in skeletal muscle biopsies from ISCU myopathy patients
[76]. These results suggest the existence of a Fe–S cluster-dependent regulation of
mitochondrial iron homeostasis. Does it mean that a particular transcription factor,
likely, regulated by a Fe–S-dependent peptide or protein, modulates mitochondrial
iron acquisition and retention [77]?
The neurological phenotypes of FRDA reflect lesions in the central nervous sys-
tem and peripheral nervous system. The major lesions are located in the dorsal root
ganglia of spinal cord and the dentate nucleus of the cerebellum [78]. The sen-
sory peripheral nerves are often affected. Optic atrophy or hearing loss is relatively
uncommon though it may be severe enough to cause deafness in approximately 1%
of patients [79, 80]. Despite normal pure tone audiometry, patients with FRDA may
have problems with speech perception [81]. Because dentate nucleus (DN) and dorsal
root ganglions (DRGs) of individuals with FRDA are severely affected tissues, they
have been extensively studied to investigate iron dysregulation by Dr. Koeppen AH
[74]. Dentate nucleus is an iron-rich cerebellar structure that shows signs of neurode-
generation in patients with FRDA [78, 82, 83]. The much smaller size of the dentate
nucleus in FRDA than in normal specimen is particularly apparent on a macro-stain
for iron [84]. It lacks the distinct gray matter ribbon of the normal nucleus, and the
intensity of the iron staining is lower than normal [82]. Further studies revealed that
ferritin detected by immunohistochemistry is most abundant in oligodendroglia of the
DN hilus and hypertrophic microglia in the atrophic DN gray matter of FRDA [85].
So far, the presence of mitochondrial iron accumulation in FRDA-affected neurons
is unclear although it is common in heart of model organisms and FRDA patients.
7 Iron Pathophysiology in Friedreich’s Ataxia 133
Despite a report of difference in the MRI signals that suggests an overall increase
of iron in the dentate nucleus of individuals with FRDA [86, 87], no difference in
iron concentrations was demonstrated using autopsies [82]. However, modification
of the expression of iron-related proteins such as transferrin receptor1, ferritins,
and ferroportin was observed, thereby suggesting a change in iron metabolism [82],
supporting that iron was relocating, likely, from dying neurons to microglia of dentate
nucleus.
Similarly, DRGs from individuals with FRDA do not show overall iron concentra-
tions above normal [88]. However, the expression of ferritin, the iron-storage protein,
increases in satellite cells surrounding affected DRG neurons and decreases in DRG
neurons [78, 88]. Meanwhile, not only the overall size reduction of subcapsular nerve
cells in the DRG, but also the more proliferated glial cells surrounding the degener-
ated neurons are observed in DRG, in consistence with the much thicker satellite layer
around neurons in patients than in normal subjects, whereas sural nerves in FRDA
showed no convincing change in ferritin and ferroportin, militating against local iron
dysmetabolism. Surprisingly, mitochondrial iron deposits have never been reported
in neurons from FRDA individuals [89] and were not observed in an inducible con-
ditional mouse model reproducing the neuronal phenotypes (PrPmice) [90]. The
results stand out in contrast to the previously reported changes in dorsal spinal roots
of patients with FRDA. Therefore, we are not sure about any indication on the pri-
mary involvement of iron dysregulation in the development of dorsal root ganglion
in FRDA patients.
pathological areas contained granular iron and iron-laden phagocytes invade and
replace sarcoplasm and myofibrils of single fibers. The further molecular evidence
was that CD68-positive inflammatory cells express cytosolic ferritin and hepcidin,
an iron-regulatory hormone [91]. The retention of iron in macrophages is a likely
contributor to heart failure in FRDA due to the highly presented hepcidin to keep
the iron from export by ferroportin. The unevenly distributed iron in heart suggested
that iron accumulation progressed from a few small granules to coarse aggregates in
phagocytized cardiomyocytes or infiltrated macrophages to result in elevated levels
of iron in highly localized, randomly distributed regions of the heart.
Altogether, these observations led to the early assumption of a pathophysiological
implication of iron-dependent pathways in FRDA. However, it is not clear why the
restricted accumulation of cardiac iron in FRDA should be much more damaging
than global iron overload and what drives the heterogeneous distribution of iron in
heart with variance from systemic iron overload.
At present, there is no any approved cure treatment for this progressive condition.
However, the efforts for a potential pharmacological treatment have been made con-
siderably in the past two decades with many potential therapeutic agents proposed
to delay disease progression (reviewed in Ref. [98]). An optimal therapy in FRDA
would consist of providing sufficient intracellular FXN to restore the physiological
functions of cells, including the gene therapy [99, 100] and drugs increasing FXN
expression [101]. Interestingly, two very recent studies have reported that bone mar-
row transplantation stimulates neural repair and ameliorates deficits in FRDA mice
[102, 103]. Here, we only take attention on iron-related therapies in mouse model
and human.
It is often observed that iron accumulation in mitochondria occurs in case of
impairment of Fe–S cluster biogenesis (reviewed in [29]). Iron accumulation was
first observed in the heart of FRDA patients [72] as a secondary effect of FXN
deficiency [55, 67]. The excess iron may play destructive effects in several cellular
functions due to its unique physiochemical properties. Interestingly, it seems still
controversial in the central neuronal system [82, 87]. One possibility could be the
ratio of Fe2+ /Fe3+ shifted as observed in Parkinson disease (PD) patients [104]. In this
mentioned study, iron ratio was reported to be shifted from 1:2 in healthy subjects to
2:1 in PD patients in substantia nigra pars compacta regions of the brain. The total
iron content measured with some common methods may not distinguish the Fe2+ and
Fe3+ . The increase in the concentration of highly reactive iron species, Fe2+ , results in
excess production of ROS via Fenton chemistry. Nevertheless, iron chelation therapy
was proposed and assessed by Cabantchik group [86]. In that study, they found that
a membrane-permeant and orally administered chelator, deferiprone (DFP), reduced
neuropathy and ataxic gait only in the youngest FRDA patients by shuttling chelated
iron from cells to transferrin to reduce brain iron accumulation without systemic
anemia. The results indicate that DFP is able to cross the blood–brain barrier. It
was commonly observed in a few clinical trials that DFP administration at low dose
was associated with a significant reduction in cardiac hypertrophy, or an abnormal
enlargement, or thickening, of the heart muscle (reviewed in [105]). The effects of
long-term treatment at low doses are suggested to be worthy to be evaluated later
on since most of the patients die of cardiac failure. However, the results were some-
how puzzling as different doses showed opposite effects, if any (reviewed in [106]).
Moreover, our previous results demonstrated that iron deficiency downregulated FXN
expression [107] and reduced aconitase activity even with low concentration of DFP
(Li, unpublished data) in cells derived from healthy controls and FRDA patients.
Therefore, the rationale is further questioned behind the use of chelation therapy in
FRDA. Larger studies with hard endpoints are still missing.
Things might get a turn for a hope. A combined therapy of a low dose of DFP with
idebenone is relatively safe and might improve neurological function and heart hyper-
trophy in a 6-month randomized, double-blind placebo-controlled study [108]. Some
novel bifunctional or multifunctional agents have been synthesized with the ability
136 K. Li
of iron chelation and monoamine oxidase inhibitory (MAOI) activities for ironing
out iron from the brain and preventing iron-induced Fenton reaction from hydroxyl
radical toxicity [109]. Though these pharmacophores as iron chelators are being
examined for treatment in other neurodegenerative diseases with regional increase
of iron levels (reviewed in [110, 111]), such as Parkinson and Alzheimer diseases,
the similar strategies are worth to be tried in the treatment of FRDA. Besides iron
chelation, the radical scavenging ability, the mitochondrial activity enhancing, and
upregulation of FXN expression may also be under consideration to be combined.
Recently, we used a mitochondria-targeted agent SS-31, a tetrapeptide with the abil-
ity to enrich in mitochondria more than 1000-folds than in other organelles within
cells by binding cardiolipin [112]. This binding is thought to maintain the mitochon-
drial structure and integrity to improve the mitochondrial function. Interestingly, not
only as observed to reduce the oxidative stress, but also increase of FXN expression
at translational levels was revealed in a model of cells derived from FRDA patients
[101]. When we used the GAA-containing mouse model [69], the FRDA pheno-
types were reversed and the similar mechanism was revealed to that observed in cell
model of FRDA (in preparation for publication). Therefore, drugs designed based
on iron chelation, even on more accurate characteristics for iron shuttling between
organelles, are not enough. Due to the important role of FXN in mitochondrial func-
tion, development of therapeutic strategies should end at targeting more pathways in
combination with a few drugs or simply end at elevating FXN protein levels. Gene
therapy and transplantation of stem cells are also recommended.
7.7 Summary
References
1. Colombo R, Carobene A (2000) Age of the intronic GAA triplet repeat expansion mutation
in Friedreich ataxia. Hum Genet 106:455–458
2. Nieto A, Hernández-Torres A, Pérez-Flores J, Montón F (2018) Depressive symptoms in
Friedreich ataxia. Int J Clin Health Psychol 18:18–26
3. Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cavalcanti F, Monros E,
Rodius F, Duclos F, Monticelli A, Zara F, Canizares J, Koutnikova H, Bidichandani SI, Gellera
C, Brice A, Trouillas P, De Michele G, Filla A, De Frutos R, Palau F, Patel PI, Di Donato S,
Mandel JL, Cocozza S, Koenig M, Pandolfo M (1996) Friedreich’s ataxia: autosomal recessive
disease caused by an intronic GAA triplet repeat expansion. Science 271:1423–1427
4. Cossee M, Durr A, Schmitt M, Dahl N, Trouillas P, Allinson P, Kostrzewa M, Nivelon-
Chevallier A, Gustavson KH, Kohlschutter A, Muller U, Mandel JL, Brice A, Koenig M,
Cavalcanti F, Tammaro A, De Michele G, Filla A, Cocozza S, Labuda M, Montermini L,
Poirier J, Pandolfo M (1999) Friedreich’s ataxia: point mutations and clinical presentation of
compound heterozygotes. Ann Neurol 45:200–206
5. De Castro M, Garcia-Planells J, Monros E, Canizares J, Vazquez-Manrique R, Vilchez JJ, Urta-
sun M, Lucas M, Navarro G, Izquierdo G, Molto MD, Palau F (2000) Genotype and phenotype
analysis of Friedreich’s ataxia compound heterozygous patients. Hum Genet 106:86–92
138 K. Li
25. Gakh O, Bedekovics T, Duncan SF, Smith DY, Berkholz DS, Isaya G (2010) Normal and
Friedreich ataxia cells express different isoforms of frataxin with complementary roles in
iron-sulfur cluster assembly. J Biol Chem 285:38486–38501
26. Musco G, Stier G, Kolmerer B, Adinolfi S, Martin S, Frenkiel T, Gibson T, Pastore A (2000)
Towards a structural understanding of Friedreich’s ataxia: the solution structure of frataxin.
Structure 8:695–707
27. Dhe-Paganon S, Shigeta R, Chi YI, Ristow M, Shoelson SE (2000) Crystal structure of human
frataxin. J Biol Chem 275:30753–30756
28. Rotig A, de Lonlay P, Chretien D, Foury F, Koenig M, Sidi D, Munnich A, Rustin P (1997)
Aconitase and mitochondrial iron-sulphur protein deficiency in Friedreich ataxia. Nat Genet
17:215–217
29. Stehling O, Lill R (2013) The role of mitochondria in cellular iron-sulfur protein biogenesis:
mechanisms, connected processes, and diseases. Cold Spring Harb Perspect Biol 5:a011312
30. Gerber J, Muhlenhoff U, Lill R (2003) An interaction between frataxin and Isu1/Nfs1 that is
crucial for Fe/S cluster synthesis on Isu1. EMBO Rep 4:906–911
31. Layer G, Ollagnier-de Choudens S, Sanakis Y, Fontecave M (2006) Iron-sulfur cluster biosyn-
thesis: characterization of Escherichia coli CYaY as an iron donor for the assembly of [2Fe–2S]
clusters in the scaffold IscU. J Biol Chem 281:16256–16263
32. Shan Y, Napoli E, Cortopassi G (2007) Mitochondrial frataxin interacts with ISD11 of the
NFS1/ISCU complex and multiple mitochondrial chaperones. Hum Mol Genet 16:929–941
33. Li H, Gakh O, Smith DY, Isaya G (2009) Oligomeric yeast frataxin drives assembly of core
machinery for mitochondrial iron-sulfur cluster synthesis. J Biol Chem 284:21971–21980
34. Alsina D, Purroy R, Ros J, Tamarit J (2018) Iron in Friedreich Ataxia: a central role in the
pathophysiology or an epiphenomenon? Pharmaceuticals (Basel) 11
35. Gakh O, Ranatunga W, Smith DY, Ahlgren EC, Al-Karadaghi S, Thompson JR, Isaya G
(2016) Architecture of the human mitochondrial iron-sulfur cluster assembly machinery. J
Biol Chem 291:21296–21321
36. Armas AM, Balparda M, Terenzi A, Busi MV, Pagani MA, Gomez-Casati DF (2019) Fer-
rochelatase activity of plant frataxin. Biochimie 156:118–122
37. Cook JD, Bencze KZ, Jankovic AD, Crater AK, Busch CN, Bradley PB, Stemmler AJ, Spaller
MR, Stemmler TL (2006) Monomeric yeast frataxin is an iron-binding protein. Biochemistry
45:7767–7777
38. Yoon T, Cowan JA (2003) Iron-sulfur cluster biosynthesis. Characterization of frataxin as
an iron donor for assembly of [2Fe–2S] clusters in ISU-type proteins. J Am Chem Soc
125:6078–6084
39. Bencze KZ, Yoon T, Millan-Pacheco C, Bradley PB, Pastor N, Cowan JA, Stemmler TL (2007)
Human frataxin: iron and ferrochelatase binding surface. Chem Commun (Camb) 1798–1800
40. Yoon T, Cowan JA (2004) Frataxin-mediated iron delivery to ferrochelatase in the final step
of heme biosynthesis. J Biol Chem 279:25943–25946
41. Steinkellner H, Singh HN, Muckenthaler MU, Goldenberg H, Moganty RR, Scheiber-
Mojdehkar B, Sturm B (2017) No changes in heme synthesis in human Friedreich s ataxia
erythroid progenitor cells. Gene 621:5–11
42. Adamec J, Rusnak F, Owen WG, Naylor S, Benson LM, Gacy AM, Isaya G (2000) Iron-
dependent self-assembly of recombinant yeast frataxin: implications for Friedreich ataxia.
Am J Hum Genet 67:549–562
43. Ahlgren EC, Fekry M, Wiemann M, Soderberg CA, Bernfur K, Gakh O, Rasmussen M,
Hojrup P, Emanuelsson C, Isaya G, Al-Karadaghi S (2017) Iron-induced oligomerization of
human FXN81-210 and bacterial CyaY frataxin and the effect of iron chelators. PLoS ONE
12:e0188937
44. Schagerlof U, Elmlund H, Gakh O, Nordlund G, Hebert H, Lindahl M, Isaya G, Al-Karadaghi
S (2008) Structural basis of the iron storage function of frataxin from single-particle recon-
struction of the iron-loaded oligomer. Biochemistry 47:4948–4954
45. Park S, Gakh O, Mooney SM, Isaya G (2002) The ferroxidase activity of yeast frataxin. J Biol
Chem 277:38589–38595
140 K. Li
46. Soderberg CA, Rajan S, Shkumatov AV, Gakh O, Schaefer S, Ahlgren EC, Svergun DI, Isaya
G, Al-Karadaghi S (2013) The molecular basis of iron-induced oligomerization of frataxin
and the role of the ferroxidation reaction in oligomerization. J Biol Chem 288:8156–8167
47. Sutak R, Seguin A, Garcia-Serres R, Oddou JL, Dancis A, Tachezy J, Latour JM, Camadro
JM, Lesuisse E (2012) Human mitochondrial ferritin improves respiratory function in yeast
mutants deficient in iron-sulfur cluster biogenesis, but is not a functional homologue of yeast
frataxin. Microbiologyopen 1:95–104
48. Vaubel RA, Isaya G (2013) Iron-sulfur cluster synthesis, iron homeostasis and oxidative stress
in Friedreich ataxia. Mol Cell Neurosci 55:50–61
49. Shaw GC, Cope JJ, Li L, Corson K, Hersey C, Ackermann GE, Gwynn B, Lambert AJ,
Wingert RA, Traver D, Trede NS, Barut BA, Zhou Y, Minet E, Donovan A, Brownlie A,
Balzan R, Weiss MJ, Peters LL, Kaplan J, Zon LI, Paw BH (2006) Mitoferrin is essential for
erythroid iron assimilation. Nature 440:96–100
50. Tsai CL, Barondeau DP (2010) Human frataxin is an allosteric switch that activates the Fe–S
cluster biosynthetic complex. Biochemistry 49:9132–9139
51. Colin F, Martelli A, Clemancey M, Latour JM, Gambarelli S, Zeppieri L, Birck C, Page A,
Puccio H, Ollagnier de Choudens S (2015) Mammalian frataxin controls sulfur production
and iron entry during de novo Fe4S4 cluster assembly. J Am Chem Soc 135:733–740
52. Bridwell-Rabb J, Fox NG, Tsai CL, Winn AM, Barondeau DP (2014) Human frataxin
activates Fe–S cluster biosynthesis by facilitating sulfur transfer chemistry. Biochemistry
53:4904–4913
53. Foury F, Cazzalini O (1997) Deletion of the yeast homologue of the human gene associated
with Friedreich’s ataxia elicits iron accumulation in mitochondria. FEBS Lett 411:373–377
54. Llorens JV, Soriano S, Calap-Quintana P, Gonzalez-Cabo P, Molto MD (2019) The role of
Iron in Friedreich’s ataxia: insights from studies in human tissues and cellular and animal
models. Front Neurosci 13:75
55. Cossee M, Puccio H, Gansmuller A, Koutnikova H, Dierich A, LeMeur M, Fischbeck K,
Dolle P, Koenig M (2000) Inactivation of the Friedreich ataxia mouse gene leads to early
embryonic lethality without iron accumulation. Hum Mol Genet 9:1219–1226
56. Babcock M, de Silva D, Oaks R, Davis-Kaplan S, Jiralerspong S, Montermini L, Pandolfo M,
Kaplan J (1997) Regulation of mitochondrial iron accumulation by Yfh1p, a putative homolog
of frataxin. Science 276:1709–1712
57. Canizares J, Blanca JM, Navarro JA, Monros E, Palau F, Molto MD (2000) dfh is a Drosophila
homolog of the Friedreich’s ataxia disease gene. Gene 256:35–42
58. Llorens JV, Navarro JA, Martinez-Sebastian MJ, Baylies MK, Schneuwly S, Botella JA,
Molto MD (2007) Causative role of oxidative stress in a Drosophila model of Friedreich
ataxia. FASEB J. 21:333–344
59. Anderson PR, Kirby K, Orr WC, Hilliker AJ, Phillips JP (2008) Hydrogen peroxide scaveng-
ing rescues frataxin deficiency in a Drosophila model of Friedreich’s ataxia. Proc Natl Acad
Sci U S A 105:611–616
60. Calap-Quintana P, Navarro JA, Gonzalez-Fernandez J, Martinez-Sebastian MJ, Molto MD,
Llorens JV (2018) Drosophila melanogaster Models of Friedreich’s Ataxia. Biomed Res Int
2018:5065190
61. Vazquez-Manrique RP, Gonzalez-Cabo P, Ortiz-Martin I, Ros S, Baylis HA, Palau F (2007)
The frataxin-encoding operon of Caenorhabditis elegans shows complex structure and regu-
lation. Genomics 89:392–401
62. Vazquez-Manrique RP, Gonzalez-Cabo P, Ros S, Aziz H, Baylis HA, Palau F (2006) Reduction
of caenorhabditis elegans frataxin increases sensitivity to oxidative stress, reduces lifespan,
and causes lethality in a mitochondrial complex II mutant. FASEB J. 20:172–174
63. Gonzalez-Cabo P, Ros S, Palau F (2010) Flavin adenine dinucleotide rescues the phenotype
of frataxin deficiency. PLoS ONE 5:e8872
64. Ventura N, Rea S, Henderson ST, Condo I, Johnson TE, Testi R (2005) Reduced expression
of frataxin extends the lifespan of Caenorhabditis elegans. Aging Cell 4:109–112
7 Iron Pathophysiology in Friedreich’s Ataxia 141
84. Koeppen AH (2011) Friedreich’s ataxia: pathology, pathogenesis, and molecular genetics. J
Neurol Sci 303:1–12
85. Koeppen AH, Ramirez RL, Yu D, Collins SE, Qian J, Parsons PJ, Yang KX, Chen Z,
Mazurkiewicz JE, Feustel PJ (2012) Friedreich’s ataxia causes redistribution of iron, cop-
per, and zinc in the dentate nucleus. Cerebellum 11:845–860
86. Boddaert N, Le Quan Sang KH, Rotig A, Leroy-Willig A, Gallet S, Brunelle F, Sidi D,
Thalabard JC, Munnich A, Cabantchik ZI (2007) Selective iron chelation in Friedreich ataxia:
biologic and clinical implications. Blood 110:401–408
87. Waldvogel D, van Gelderen P, Hallett M (1999) Increased iron in the dentate nucleus of
patients with Friedrich’s ataxia. Ann Neurol 46:123–125
88. Koeppen AH, Morral JA, Davis AN, Qian J, Petrocine SV, Knutson MD, Gibson WM,
Cusack MJ, Li D (2009) The dorsal root ganglion in Friedreich’s ataxia. Acta Neuropathol
118:763–776
89. Morral JA, Davis AN, Qian J, Gelman BB, Koeppen AH (2010) Pathology and pathogenesis
of sensory neuropathy in Friedreich’s ataxia. Acta Neuropathol 120:97–108
90. Simon D, Seznec H, Gansmuller A, Carelle N, Weber P, Metzger D, Rustin P, Koenig M,
Puccio H (2004) Friedreich ataxia mouse models with progressive cerebellar and sensory
ataxia reveal autophagic neurodegeneration in dorsal root ganglia. J Neurosci 24:1987–1995
91. Koeppen AH, Ramirez RL, Becker AB, Bjork ST, Levi S, Santambrogio P, Parsons PJ, Kruger
PC, Yang KX, Feustel PJ, Mazurkiewicz JE (2015) The pathogenesis of cardiomyopathy in
Friedreich ataxia. PLoS ONE 10:e0116396
92. Michael S, Petrocine SV, Qian J, Lamarche JB, Knutson MD, Garrick MD, Koeppen AH
(2006) Iron and iron-responsive proteins in the cardiomyopathy of Friedreich’s ataxia. Cere-
bellum 5:257–267
93. Kruger PC, Yang KX, Parsons PJ, Becker AB, Feustel PJ, Koeppen AH (2016) Abundance
and significance of Iron, Zinc, Copper, and Calcium in the hearts of patients with Friedreich
Ataxia. Am J Cardiol 118:127–131
94. Garg M, Kulkarni SD, Shah KN, Hegde AU (2017) Diabetes Mellitus as the Presenting Feature
of Friedreich’s Ataxia. J Neurosci Rural Pract 8:S117–S119
95. Cnop M, Igoillo-Esteve M, Rai M, Begu A, Serroukh Y, Depondt C, Musuaya AE, Marhfour
I, Ladriere L, Moles Lopez X, Lefkaditis D, Moore F, Brion JP, Cooper JM, Schapira AH,
Clark A, Koeppen AH, Marchetti P, Pandolfo M, Eizirik DL, Fery F (2012) Central role and
mechanisms of beta-cell dysfunction and death in friedreich ataxia-associated diabetes. Ann
Neurol 72:971–982
96. Del Guerra S, D’Aleo V, Gualtierotti G, Pandolfi R, Boggi U, Vistoli F, Barnini S, Filipponi
F, Del Prato S, Lupi R (2012) Evidence for a role of frataxin in pancreatic islets isolated from
multi-organ donors with and without type 2 diabetes mellitus. Horm Metab Res 44:471–475
97. Igoillo-Esteve M, Gurgul-Convey E, Hu A, Romagueira Bichara Dos Santos L, Abdulkarim
B, Chintawar S, Marselli L, Marchetti P, Jonas JC, Eizirik DL, Pandolfo M, Cnop M (2015)
Unveiling a common mechanism of apoptosis in beta-cells and neurons in Friedreich’s ataxia.
Hum Mol Genet 24:2274–2286
98. Tai G, Corben LA, Yiu EM, Milne SC, Delatycki MB (2018) Progress in the treatment of
Friedreich ataxia. Neurol Neurochir Pol 52:129–139
99. Perdomini M, Belbellaa B, Monassier L, Reutenauer L, Messaddeq N, Cartier N, Crystal RG,
Aubourg P, Puccio H (2014) Prevention and reversal of severe mitochondrial cardiomyopathy
by gene therapy in a mouse model of Friedreich’s ataxia. Nat Med 20:542–547
100. Piguet F, de Montigny C, Vaucamps N, Reutenauer L, Eisenmann A, Puccio H (2018) Rapid
and complete reversal of sensory Ataxia by gene therapy in a novel model of Friedreich
Ataxia. Mol Ther 26:1940–1952
101. Zhao H, Li H, Hao S, Chen J, Wu J, Song C, Zhang M, Qiao T, Li K (2017) Peptide SS-31
upregulates frataxin expression and improves the quality of mitochondria: implications in the
treatment of Friedreich ataxia. Sci Rep 7:9840
102. Rocca CJ, Goodman SM, Dulin JN, Haquang JH, Gertsman I, Blondelle J, Smith JLM, Heyser
CJ, Cherqui S (2017) Transplantation of wild-type mouse hematopoietic stem and progenitor
cells ameliorates deficits in a mouse model of Friedreich’s ataxia. Sci Transl Med 9
7 Iron Pathophysiology in Friedreich’s Ataxia 143
103. Kemp KC, Hares K, Redondo J, Cook AJ, Haynes HR, Burton BR, Pook MA, Rice CM, Scold-
ing NJ, Wilkins A (2018) Bone marrow transplantation stimulates neural repair in Friedreich’s
ataxia mice. Ann Neurol 83:779–793
104. Riederer P, Rausch WD, Schmidt B, Kruzik P, Konradi C, Sofic E, Danielczyk W, Fischer M,
Ogris E (1988) Biochemical fundamentals of Parkinson’s disease. Mt Sinai J Med 55:21–28
105. Nunez MT, Chana-Cuevas P (2018) New perspectives in iron chelation therapy for the treat-
ment of neurodegenerative diseases. Pharmaceuticals (Basel) 11
106. Pandolfo M, Hausmann L (2013) Deferiprone for the treatment of Friedreich’s ataxia. J
Neurochem 126(Suppl 1):142–146
107. Li K, Besse EK, Ha D, Kovtunovych G, Rouault TA (2008) Iron-dependent regulation
of frataxin expression: implications for treatment of Friedreich ataxia. Hum Mol Genet
17:2265–2273
108. Elincx-Benizri S, Glik A, Merkel D, Arad M, Freimark D, Kozlova E, Cabantchik I, Hassin-
Baer S (2016) Clinical experience with deferiprone treatment for Friedreich Ataxia. J Child
Neurol 31:1036–1040
109. Youdim MB, Fridkin M, Zheng H (2004) Novel bifunctional drugs targeting monoamine
oxidase inhibition and iron chelation as an approach to neuroprotection in Parkinson’s disease
and other neurodegenerative diseases. J Neural Transm (Vienna) 111:1455–1471
110. Singh YP, Pandey A, Vishwakarma S, Modi G (2018) A review on iron chelators as potential
therapeutic agents for the treatment of Alzheimer’s and Parkinson’s diseases. Mol Divers
111. Youdim MB, Fridkin M, Zheng H (2005) Bifunctional drug derivatives of MAO-B inhibitor
rasagiline and iron chelator VK-28 as a more effective approach to treatment of brain ageing
and ageing neurodegenerative diseases. Mech Ageing Dev 126:317–326
112. Szeto HH (2014) First-in-class cardiolipin-protective compound as a therapeutic agent to
restore mitochondrial bioenergetics. Br J Pharmacol 171:2029–2050
113. Myers L, Farmer JM, Wilson RB, Friedman L, Tsou A, Perlman SL, Subramony SH, Gomez
CM, Ashizawa T, Wilmot GR, Mathews KD, Balcer LJ, Lynch DR (2008) Antioxidant use
in Friedreich ataxia. J Neurol Sci 267:174–176
Chapter 8
The Role of Iron in Amyotrophic Lateral
Sclerosis
8.1 Introduction
X.-L. Bu
Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military
Medical University, Chongqing, China
Y. Xiang
Department of Neurology, Chengdu Military General Hospital, Chengdu, China
Y. Guo (B)
Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing, China
e-mail: [email protected]
© Springer Nature Singapore Pte Ltd. 2019 145
Y.-Z. Chang (ed.), Brain Iron Metabolism and CNS Diseases,
Advances in Experimental Medicine and Biology 1173,
https://doi.org/10.1007/978-981-13-9589-5_8
146 X.-L. Bu et al.
suggest that redox metals, mitochondrial dysfunction, oxidative stress, and neuroin-
flammation are involved in the loss of motor neurons [2–4].
Iron has been proved to play a crucial role in oxygen transportation, cell prolifera-
tion and differentiation, DNA synthesis, myelin production, synthesis of neurotrans-
mitters, and the generation of oxidative stress [5]. Many studies indicate that iron
overload may be involved in ALS pathogenesis. In this review, we summarize the
current knowledge of iron metabolism disturbances and its pathogenic role in ALS,
and discuss the recent progress and problems with respect to iron chelator therapy
on ALS, aiming to propose a new therapeutic strategy to ameliorate the progression
of the disease.
Transferrin receptor 1 (TFR1) is responsible for the endocytosis of the iron uptake,
and it is highly expressed on the luminal side of endothelial cells [6]. Blood iron enters
the endothelial cells of the blood–brain barrier via TFR1-mediated endocytosis [7–9].
Iron could also be released into the extracellular environment and then be taken up
by neurons, microglia, and astrocytes [5, 10].
Neurons are thought to uptake iron via the transferrin receptors and divalent metal
transporter 1 (DMT 1). Meanwhile, neurons express ferroportin, which is involved
in the efflux of iron [11]. It has been shown that inflammatory stimuli could increase
DMT1 expression while decrease the expression of ferroportin [12], causing the iron
accumulation in neurons. Astrocyte, the important component of the blood-brain
barrier, plays a key role in regulating brain iron absorption [13]. Although astrocytes
do not express TFR, it is able to take up iron via the DMT1 that expressed in the
perivascular end-foot processes of astrocytes [14]. The glycosylphosphatidylinositol-
anchored form of ceruloplasmin expressed by astrocytes plays a role in oxidizing
ferrous iron, and could help iron efflux from cells [15]. It shows that lack of cerulo-
plasmin can lead to iron accumulation in the brain and neurodegeneration [16]. Iron
is required for axon myelination in oligodendrocytes. It has been proved that oligo-
dendrocytes can take up interstitial ferritin via the ferritin receptor. On the other hand,
oligodendrocyte is able to uptake iron from blood vessels [5]. Inflammatory stim-
uli increases iron accumulation in microglia by increasing the expression of DMT1
protein [17, 18]. It showed that iron uptake was increased and iron exportation was
down-regulated when the microglia is activated [19]. Moreover, inflammatory stim-
uli could increase hepcidin concentrations in astrocytes and microglia, and decrease
ferroportin level in astrocytes, microglia, and neurons [12].
Neuroinflammation is a chronic activation of the immune response in the central
nervous system and related to neuronal dysfunction in neurodegenerative diseases.
Microglia could be switched from their normal quiescent state to an activated state by
the systemic inflammation with aging [20]. Age-dependent enhanced neuroinflam-
matory processes in the brain refer to increased microglia and astrocyte numbers
and proinflammatory cytokines secreted by activated glial cells with aging [20]. Iron
8 The Role of Iron in Amyotrophic Lateral Sclerosis 147
levels in the substantia nigra, putamen, globus pallidus, caudate nucleus, and cortices
increase with age [5]. Additionally, the blood–brain barrier permeability increases
in the process of aging. Therefore, brain iron load may increase during aging.
Numerous studies have examined the iron levels in ALS patients. A prior study
reported in 1985 showed that the iron concentrations of erythrocytes were the same
between ALS and controls [21]. Goodall et al. declared that the serum ferritin levels
were significantly increased in ALS patients, while no significant differences in the
concentrations of iron, transferrin, iron saturation, or total iron binding capacity were
found [22]. It also reported that ALS patients had significantly higher plasma levels
of ferritin but lower concentrations of transferrin [23, 24], and the patients with a
higher ferritin level had a shorter survival time [25]. A recent study firstly showed
that the serum iron, ferritin, and transferrin saturation coefficient were significantly
elevated in ALS patients, and the iron status was associated with body weight loss
[26]. And serum ferritin may be a candidate biomarker for ALS aggravation [27]. Free
iron level was also increased in the cerebrospinal fluid of ALS patients compared
to controls [28, 29]. And twofold increased level of inappropriate iron ligands in
cerebrospinal fluid (CSF) are found in patients with ALS, which may increase iron
redox activity and reactive oxygen species (ROS) production [30].
Autopsy study demonstrated that iron load in gray matter from the frontal cortex
of ALS was increased significantly than that of controls [31]. The concentration of
iron was also increased in the spinal cord of ALS patients [32–34]. Disruption in
the expression of brain iron transporters such as lactotransferrin receptor, melan-
otransferrin, and ceruloplasmin is related to iron accumulation in ALS [35]. Iron
accumulation could also be observed in the motor cortex using magnetic resonance
imaging (MRI), which appears to be a very promising biomarker for ALS [36, 37].
MRI also found showed increased iron deposits in the globus pallidus of frontotem-
poral dementia with ALS [38]. 7T MRI showed that ALS patients had motor cortex
hypointensity, and the postmortem studies showed that the increased iron accumu-
lation in microglial cells may be responsible for the location of the signal changes
on MRI [39]. Recent researches confirmed that iron level was significantly higher in
the motor cortex of ALS patients by susceptibility-weighted imaging (SWI), which
may be useful biomarkers for the diagnosis of clinically suspected ALS [40, 41].
148 X.-L. Bu et al.
Iron is a redox ion and can aggravate oxidative stress by generating ROS, which
may lead to neuronal cell damage, aging, and apoptosis. It demonstrated that the
increased iron load can form redox active complexes which participates in the Fen-
ton reaction and produces the toxic hydroxyl radical [42–44]. Meanwhile, ROS can
induce the release of iron from the iron storage proteins such as mitochondrial iron—
sulfur cluster proteins [45]. Increased oxidative stress occurred in ALS could also
be promoted by the genetic abnormalities (e.g., superoxide dismutase mutations)
[46]. Iron accumulation also increases the enzymatic activity of TNF-α converting
enzyme, leading to secretion of TNF-α and promoting neuroinflammation [47]. A
number of studies have demonstrated that mutations of genes encoding the proteins
involved in iron regulation are related to neuronal cell death also in ALS [48, 49]. The
mutation of HFE gene, which encoding protein interacts with the transferrin receptor
and regulates transferrin-mediated iron uptake, altered brain iron profiles, increased
oxidative stress, and accelerated disease process in mice [50, 51]. The mutation of
SLC11A2 gene that encoding DMT1 is associated with shorter disease duration in
ALS cases [52]. On the other hand, activation of genes involved in iron homeostasis
can induce ferritin overexpression and thus limit toxic hydroxyl radical and neuronal
loss [53]. Thus, iron misregulation promotes oxidative stress, which disrupts cellular
iron balance and causes progressive loss of motor neurons and consequent oxidative
stress, thus giving rise to a vicious circle.
In view of the key role of iron in the pathogenesis of ALS, researchers explore the
efficacy and therapeutic potential of brain-permeable iron-chelating drugs for ALS in
animal models. An earlier study found that intraperitoneal injection of iron porphyrin
significantly decreased neuronal death and improved motor function in G93A mutant
SOD1 transgenic mouse model of ALS [54]. VAR10303, the brain-permeable iron-
chelating radical scavenging drug, exerted several beneficial effects in SOD1G93A
transgenic ALS mice, including alleviating the iron accumulation and motoneuron
loss in the spinal cord, improving motor performance and prolonging survival time
[55]. Iron-chelating drugs M30 and HLA20 showed neuroprotective effects against
oxidative stress and can extend the survival time of ALS transgenic mice [56]. Iron
chelators VK-28 also significantly alleviated the increased levels of iron and oxygen
free radicals and suppressed glial activation in the spinal cords of the ALS mice
[57]. What’s more, it was able to decrease TDP-43 protein aggregation and the
proapoptotic molecule Bax and to increase the expression of antiapoptotic protein
Bcl-2 [57]. A recent pilot clinical trial showed that deferiprone could remarkably
decrease the iron levels in the cervical spinal cord, medulla oblongata, and motor
8 The Role of Iron in Amyotrophic Lateral Sclerosis 149
cortex, the levels of oxidative stress markers and neurofilament light chains in the
CSF, and attenuate the disability progression and weight loss in patients with ALS
[58]. Therefore, brain-permeable iron-chelating drugs hold a promise as therapeutic
agents for ALS by targeting multiple key pathways of the disease pathogenesis. And
the effectiveness of iron chelators in ALS therapy supports the role of iron in the
ALS pathogenesis.
8.6 Conclusion
In summary, abundant evidence shows that iron accumulation in motor cortex and
spinal cord is a hallmark of ALS, and increased iron load, in particular, in regions of
central nervous system causes neuronal cell damage. However, it is uncertain whether
iron accumulation is a primary cause of ALS or consequential pathological change.
And why iron accumulation in ALS is selective for some brain areas is also unclear.
To further clarify the pathophysiological role of iron in ALS pathogenesis, future
studies should focus on the chemical state of iron in healthy control and ALS patients,
and its specifically pathogenic role in ALS different from other neurodegenerative
disorders.
More randomized, double-blind, placebo-controlled, multicenter trials are needed
to evaluate the effect of iron chelators on patients with ALS.
References
1. Vandoorne T, De Bock K, Van Den Bosch L (2018) Energy metabolism in ALS: an underap-
preciated opportunity? Acta Neuropathol 135:489–509
2. Carri MT, Ferri A, Cozzolino M, Calabrese L, Rotilio G (2003) Neurodegeneration in amy-
otrophic lateral sclerosis: the role of oxidative stress and altered homeostasis of metals. Brain
Res Bull 61:365–374
3. Hadzhieva M, Kirches E, Wilisch-Neumann A, Pachow D, Wallesch M, Schoenfeld P, Paege
I, Vielhaber S, Petri S, Keilhoff G, Mawrin C (2013) Dysregulation of iron protein expression
in the G93A model of amyotrophic lateral sclerosis. Neuroscience 230:94–101
4. Tokuda E, Furukawa Y (2016) Copper homeostasis as a therapeutic target in amyotrophic
lateral sclerosis with SOD1 mutations. Int J Mol Sci 17(5):636
5. Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L (2014) The role of iron in brain ageing
and neurodegenerative disorders. Lancet Neurol 13:1045–1060
6. Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY (1984) Transferrin
receptor on endothelium of brain capillaries. Nature 312:162–163
7. Moos T, Rosengren Nielsen T, Skjorringe T, Morgan EH (2007) Iron trafficking inside the
brain. J Neurochem 103:1730–1740
8. Ke Y, Qian ZM (2007) Brain iron metabolism: neurobiology and neurochemistry. Prog Neu-
robiol 83:149–173
9. Leitner DF, Connor JR (1820) Functional roles of transferrin in the brain. Biochim Biophys
Acta 2012:393–402
10. Biasiotto G, Di Lorenzo D, Archetti S, Zanella I (2016) Iron and neurodegeneration: is fer-
ritinophagy the link? Mol Neurobiol 53:5542–5574
150 X.-L. Bu et al.
11. Boserup MW, Lichota J, Haile D, Moos T (2011) Heterogenous distribution of ferroportin-
containing neurons in mouse brain. Biometals 24:357–375
12. Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP, Arredondo M, Gonzalez-Billault C, Nunez
MT (2013) Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes
iron accumulation in central nervous system cells. J Neurochem 126:541–549
13. Dringen R, Bishop GM, Koeppe M, Dang TN, Robinson SR (2007) The pivotal role of astro-
cytes in the metabolism of iron in the brain. Neurochem Res 32:1884–1890
14. Abbott NJ, Ronnback L, Hansson E (2006) Astrocyte-endothelial interactions at the blood-
brain barrier. Nat Rev Neurosci 7:41–53
15. Patel BN, David S (1997) A novel glycosylphosphatidylinositol-anchored form of ceruloplas-
min is expressed by mammalian astrocytes. J Biol Chem 272:20185–20190
16. Jeong SY, David S (2003) Glycosylphosphatidylinositol-anchored ceruloplasmin is required
for iron efflux from cells in the central nervous system. J Biol Chem 278:27144–27148
17. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, Ganz T (2004) IL-6 medi-
ates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone
hepcidin. J Clin Invest 113:1271–1276
18. Lee P, Peng H, Gelbart T, Beutler E (2004) The IL-6- and lipopolysaccharide-induced tran-
scription of hepcidin in HFE-, transferrin receptor 2-, and beta 2-microglobulin-deficient hep-
atocytes. Proc Natl Acad Sci U S A 101:9263–9265
19. Xu H, Wang Y, Song N, Wang J, Jiang H, Xie J (2017) New progress on the role of glia in iron
metabolism and iron-induced degeneration of dopamine neurons in Parkinson’s disease. Front
Mol Neurosci 10:455
20. Liu YH, Zeng F, Wang YR, Zhou HD, Giunta B, Tan J, Wang YJ (2013) Immunity and
Alzheimer’s disease: immunological perspectives on the development of novel therapies. Drug
Discovery Today 18:1212–1220
21. Nagata H, Miyata S, Nakamura S, Kameyama M, Katsui Y (1985) Heavy metal concentrations
in blood cells in patients with amyotrophic lateral sclerosis. J Neurol Sci 67:173–178
22. Goodall EF, Haque MS, Morrison KE (2008) Increased serum ferritin levels in amyotrophic
lateral sclerosis (ALS) patients. J Neurol 255:1652–1656
23. Mitchell RM, Simmons Z, Beard JL, Stephens HE, Connor JR (2010) Plasma biomarkers
associated with ALS and their relationship to iron homeostasis. Muscle Nerve 42:95–103
24. Qureshi M, Brown RH Jr, Rogers JT, Cudkowicz ME (2008) Serum ferritin and metal levels
as risk factors for amyotrophic lateral sclerosis. Open Neurol J 2:51–54
25. Nadjar Y, Gordon P, Corcia P, Bensimon G, Pieroni L, Meininger V, Salachas F (2012) Elevated
serum ferritin is associated with reduced survival in amyotrophic lateral sclerosis. PLoS ONE
7:e45034
26. Veyrat-Durebex C, Corcia P, Mucha A, Benzimra S, Mallet C, Gendrot C, Moreau C, Devos
D, Piver E, Pages JC, Maillot F, Andres CR, Vourc’h P, Blasco H (2014) Iron metabolism
disturbance in a French cohort of ALS patients. Biomed Res Int 2014:485723
27. Yu J, Wang N, Qi F, Wang X, Zhu Q, Lu Y, Zhang H, Che F, Li W (2018) Serum ferritin is
a candidate biomarker of disease aggravation in amyotrophic lateral sclerosis. Biomed Rep
9:333–338
28. Kokic AN, Stevic Z, Stojanovic S, Blagojevic DP, Jones DR, Pavlovic S, Niketic V, Apostolski
S, Spasic MB (2005) Biotransformation of nitric oxide in the cerebrospinal fluid of amyotrophic
lateral sclerosis patients. Redox Rep 10:265–270
29. Hozumi I, Hasegawa T, Honda A, Ozawa K, Hayashi Y, Hashimoto K, Yamada M, Koumura
A, Sakurai T, Kimura A, Tanaka Y, Satoh M, Inuzuka T (2011) Patterns of levels of biological
metals in CSF differ among neurodegenerative diseases. J Neurol Sci 303:95–99
30. Ignjatovic A, Stevic Z, Lavrnic D, Nikolic-Kokic A, Blagojevic D, Spasic M, Spasojevic I
(2012) Inappropriately chelated iron in the cerebrospinal fluid of amyotrophic lateral sclerosis
patients. Amyotroph Lateral Scler 13:357–362
31. Yasui M, Ota K, Garruto RM (1993) Concentrations of zinc and iron in the brains of Guama-
nian patients with amyotrophic lateral sclerosis and parkinsonism-dementia. Neurotoxicology
14:445–450
8 The Role of Iron in Amyotrophic Lateral Sclerosis 151
32. Ince PG, Shaw PJ, Candy JM, Mantle D, Tandon L, Ehmann WD, Markesbery WR (1994) Iron,
selenium and glutathione peroxidase activity are elevated in sporadic motor neuron disease.
Neurosci Lett 182:87–90
33. Kasarskis EJ, Tandon L, Lovell MA, Ehmann WD (1995) Aluminum, calcium, and iron in the
spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe mass
spectroscopy: a preliminary study. J Neurol Sci 130:203–208
34. Markesbery WR, Ehmann WD, Candy JM, Ince PG, Shaw PJ, Tandon L, Deibel MA (1995)
Neutron activation analysis of trace elements in motor neuron disease spinal cord. Neurode-
generation 4:383–390
35. Qian ZM, Wang Q (1998) Expression of iron transport proteins and excessive iron accumulation
in the brain in neurodegenerative disorders. Brain Res Brain Res Rev 27:257–267
36. Ignjatovic A, Stevic Z, Lavrnic S, Dakovic M, Bacic G (2013) Brain iron MRI: a biomarker
for amyotrophic lateral sclerosis. J Magn Reson Imaging 38:1472–1479
37. Oba H, Araki T, Ohtomo K, Monzawa S, Uchiyama G, Koizumi K, Nogata Y, Kachi K,
Shiozawa Z, Kobayashi M (1993) Amyotrophic lateral sclerosis: T2 shortening in motor cortex
at MR imaging. Radiology 189:843–846
38. Santillo AF, Skoglund L, Lindau M, Eeg-Olofsson KE, Tovi M, Engler H, Brundin RM, Ing-
vast S, Lannfelt L, Glaser A, Kilander L (2009) Frontotemporal dementia-amyotrophic lateral
sclerosis complex is simulated by neurodegeneration with brain iron accumulation. Alzheimer
Dis Assoc Disord 23:298–300
39. Kwan JY, Jeong SY, Van Gelderen P, Deng HX, Quezado MM, Danielian LE, Butman JA, Chen
L, Bayat E, Russell J, Siddique T, Duyn JH, Rouault TA, Floeter MK (2012) Iron accumulation
in deep cortical layers accounts for MRI signal abnormalities in ALS: correlating 7 tesla MRI
and pathology. PLoS ONE 7:e35241
40. Yu J, Qi F, Wang N, Gao P, Dai S, Lu Y, Su Q, Du Y, Che F (2014) Increased iron level in motor
cortex of amyotrophic lateral sclerosis patients: an in vivo MR study. Amyotroph Lateral Scler
Frontotemporal Degener 15:357–361
41. Adachi Y, Sato N, Saito Y, Kimura Y, Nakata Y, Ito K, Kamiya K, Matsuda H, Tsukamoto T,
Ogawa M (2015) Usefulness of SWI for the detection of iron in the motor cortex in amyotrophic
lateral sclerosis. J Neuroimaging 25:443–451
42. Popovic-Bijelic A, Mojovic M, Stamenkovic S, Jovanovic M, Selakovic V, Andjus P, Bacic
G (2016) Iron-sulfur cluster damage by the superoxide radical in neural tissues of the
SOD1(G93A) ALS rat model. Free Radic Biol Med 96:313–322
43. Sheykhansari S, Kozielski K, Bill J, Sitti M, Gemmati D, Zamboni P, Singh AV (2018) Redox
metals homeostasis in multiple sclerosis and amyotrophic lateral sclerosis: a review. Cell Death
Dis 9:348
44. Bozzo F, Mirra A, Carri MT (2017) Oxidative stress and mitochondrial damage in the patho-
genesis of ALS: new perspectives. Neurosci Lett 636:3–8
45. Horowitz MP, Greenamyre JT (2010) Mitochondrial iron metabolism and its role in neurode-
generation. J Alzheimers Dis 20(Suppl 2):S551–S568
46. Mattson MP (2004) Metal-catalyzed disruption of membrane protein and lipid signaling in the
pathogenesis of neurodegenerative disorders. Ann N Y Acad Sci 1012:37–50
47. Lee JK, Shin JH, Gwag BJ, Choi EJ (2015) Iron accumulation promotes TACE-mediated TNF-
alpha secretion and neurodegeneration in a mouse model of ALS. Neurobiol Dis 80:63–69
48. Sirabella R, Valsecchi V, Anzilotti S, Cuomo O, Vinciguerra A, Cepparulo P, Brancaccio P,
Guida N, Blondeau N, Canzoniero LMT, Franco C, Amoroso S, Annunziato L, Pignataro G
(2018) Ionic homeostasis maintenance in ALS: focus on new therapeutic targets. Front Neurosci
12:510
49. Gajowiak A, Stys A, Starzynski RR, Staron R, Lipinski P (2016) Misregulation of iron home-
ostasis in amyotrophic lateral sclerosis. Postepy Hig Med Dosw (Online) 70:709–721
50. Nandar W, Neely EB, Unger E, Connor JR (1832) A mutation in the HFE gene is associated
with altered brain iron profiles and increased oxidative stress in mice. Biochim Biophys Acta
2013:729–741
152 X.-L. Bu et al.
51. Nandar W, Neely EB, Simmons Z, Connor JR (1842) H63D HFE genotype accelerates dis-
ease progression in animal models of amyotrophic lateral sclerosis. Biochim Biophys Acta
2014:2413–2426
52. Blasco H, Vourc’h P, Nadjar Y, Ribourtout B, Gordon PH, Guettard YO, Camu W, Praline
J, Meininger V, Andres CR, Corcia P (2011) Association between divalent metal transport 1
encoding gene (SLC11A2) and disease duration in amyotrophic lateral sclerosis. J Neurol Sci
303:124–127
53. Olsen MK, Roberds SL, Ellerbrock BR, Fleck TJ, McKinley DK, Gurney ME (2001) Disease
mechanisms revealed by transcription profiling in SOD1-G93A transgenic mouse spinal cord.
Ann Neurol 50:730–740
54. Wu AS, Kiaei M, Aguirre N, Crow JP, Calingasan NY, Browne SE, Beal MF (2003) Iron
porphyrin treatment extends survival in a transgenic animal model of amyotrophic lateral
sclerosis. J Neurochem 85:142–150
55. Golko-Perez S, Amit T, Bar-Am O, Youdim MB, Weinreb O (2017) A novel iron chelator-
radical scavenger ameliorates motor dysfunction and improves life span and mitochondrial
biogenesis in SOD1(G93A) ALS Mice. Neurotox Res 31:230–244
56. Kupershmidt L, Weinreb O, Amit T, Mandel S, Carri MT, Youdim MB (2009) Neuroprotective
and neuritogenic activities of novel multimodal iron-chelating drugs in motor-neuron-like NSC-
34 cells and transgenic mouse model of amyotrophic lateral sclerosis. FASEB J 23:3766–3779
57. Wang Q, Zhang X, Chen S, Zhang X, Zhang S, Youdium M, Le W (2011) Prevention of motor
neuron degeneration by novel iron chelators in SOD1(G93A) transgenic mice of amyotrophic
lateral sclerosis. Neurodegener Dis 8:310–321
58. Moreau C, Danel V, Devedjian JC, Grolez G, Timmerman K, Laloux C, Petrault M, Gouel F,
Jonneaux A, Dutheil M, Lachaud C, Lopes R, Kuchcinski G, Auger F, Kyheng M, Duhamel
A, Perez T, Pradat PF, Blasco H, Veyrat-Durebex C, Corcia P, Oeckl P, Otto M, Dupuis L,
Garcon G, Defebvre L, Cabantchik ZI, Duce J, Bordet R, Devos D (2018) Could conservative
iron chelation lead to neuroprotection in amyotrophic lateral sclerosis? Antioxid Redox Signal
29:742–748
Chapter 9
Iron Pathophysiology
in Neurodegeneration with Brain Iron
Accumulation
9.1 Introduction
S. Levi (B)
Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University,
20132 Milan, Italy
e-mail: [email protected]
A. Cozzi · P. Santambrogio
Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
specific brain areas, mainly in the basal ganglia, even if this feature is not present in
all patients [71]. The brain iron burden has been visualized in vivo using MRI [30],
and in several cases, it was also confirmed by a histopathological examinations in
postmortem brain samples [64]. Indeed, MRI is the main method used to analyze
areas of iron accumulation in the brain, and this feature, combined with the variations
in the structures of certain areas, is the basis of distinguishing between types of NBIA
[79]. Thus, the radiological evidence of iron accumulation together with the clinical
symptoms is the preliminary diagnostic tool that suggests to clinicians to undertake
a screening to identify the specific causative gene [99]. Currently, 15 genes have
been associated with different subtypes of NBIA, the main characteristics of which
are described in Table 9.1. Among these diseases, only two, neuroferritinopathy and
aceruloplasminemia, are linked to mutations in genes that are directly involved in iron
metabolism, FTL1 and CP, respectively. All the other forms (PKAN, CoPAN, PLAN,
MPAN, FAHN, BPAN, leukoencephalopathy with dystonia and motor neuropa-
thy, Kufor–Rakeb syndrome, Woodhouse–Sakati syndrome, and GTPBP2-, CRAT-,
REPS1-, and AP4M1-associated neurodegeneration) are characterized by altered
iron metabolism and its accumulation in the brain via an as-yet-unidentified mech-
anism, possibly implicating other pathways. These disorders are indeed caused by
mutations in genes linked to lipid metabolism (PLAN, FAHN and leukoencephalopa-
thy with dystonia and motor neuropathy), mitochondrial activities (PKAN, CoPAN,
MPAN and CRAT-associated neurodegeneration), autophagy (BPAN, Kufor–Rakeb
syndrome and REPS1- and AP4M1-associated neurodegeneration) and some still
unknown functions (Woodhouse–Sakati syndrome, GTPBP2-associated neurode-
generation), all of which are apparently unrelated to iron metabolism (Table 9.1).
Interestingly, numerous genes associated with inherited NBIA encode proteins
localized in the mitochondria (PANK2, COPAN, PLA2G6, C19orf12, ATP13A2,
and CRAT). Thus, these pathologies represent an important opportunity to study and
understand the molecular mechanisms involved in brain iron management and the
connections between mitochondria function and neurodegeneration. In this review,
we focus on the reported data regarding iron alterations, with the aim of highlighting
the knowledge of the involvement of this metal in the pathogenesis of NBIA disorders.
9.2.1 Aceruloplasminemia
9.2.2 Neuroferritinopathy
Fig. 9.1 Schematic representation of L-ferritin sequence and alignment of NF variants. Human
L-ferritin sequence is matched with a schematic diagram of protein secondary structure elements
that include five α-helices segments (blue cylinders), conventionally named with letters from A to
E, connected by four loop elements and of two additional short amino acid sequences at the N and C
termini. The blue rectangle indicates the sequence interval involved in the so far detected frameshift
variants. Frameshift mutations are responsible for amino acidic changes at the C-terminus starting
from different sequence position depending on the duplication starting point. In the lower panel, the
alignment of the frameshift altered regions compared with the wild-type FtL sequence is shown.
Dashes correspond to unchanged position, while modified residues are defined
Other cases may not yet have been identified; however, NF is a very rare disorder
and thus the prevalence is not able to be calculated.
The c.460dupA mutation has been detected in the largest number of cases, 41 of
which have been carefully described [18]. The mean age of onset was approximately
40 years, and the majority of patients presented with focal onset chorea or focal dys-
tonia and reported a family history of movement disorders. The serum ferritin level
was generally low. MR imaging was abnormal in all cases. The disease progressed
relentlessly and became generalized in 5–10 years, leading to aphonia, dysphagia,
and motor disability, with cognitive dysfunction as a late feature. Characteristic facial
dystonia was common. Signs of parkinsonism, dementia, postural tremor, and cere-
bellar ataxia were also described in patients with other types of mutations [26, 86].
In general, the disorders appear to exhibit full penetrance at the age of 60 years,
with heterogeneous clinical features associated with early movement disorders fol-
lowed by minor cognitive impairments. The major and common feature is MRI signs,
with similar abnormalities observed in all affected patients. In a comparison of the
different NBIA disorders, NF was characterized by lesions in the globus pallidus,
putamen, and dentate nuclei that differed from the other NBIA subtypes. Interest-
ingly, two patients with NF showed the eye-of-the-tiger sign, which is considered
pathognomonic almost exclusively for PKAN [78].
Brain histopathology has been performed on two patients with a c.460dupA muta-
tion [22], one with a c.442dupC mutation [76, 91] and one with a c.497_498dup
160 S. Levi et al.
Two forms of NBIA are due to mutations in genes involved in the first and last steps of
the same biosynthetic pathway that produces Coenzyme A (CoA). Mutations in these
two genes, PANK2 and COASY, cause the NBIA subtypes known as pantothenate
kinase-associated neurodegeneration (PKAN, OMIM#234200) and CoA synthase
protein-associated neurodegeneration (CoPAN, OMIM*609855), respectively.
PKAN approximately accounts for 50% of all NBIA cases and, depending on
the age of onset and the rate of progression, is classified into two forms: the clas-
sical and the atypical forms. CoPAN appears to be more rare, as it has only been
identified in five subjects to date [3, 31, 33]. These two disorders display strikingly
similar phenotypes, presenting with early-onset spastic–dystonic paraparesis with a
later appearance of parkinsonian features, cognitive impairment, obsessive–compul-
sive disorder, and brain iron accumulation [31]. PKAN usually manifests in early
childhood as gait disturbances and rapidly progresses to a severe movement deficit
with dystonia, dysarthria, and dysphagia [68]. The hallmark of this disease is the
eye-of-the-tiger signal in the globus pallidus (GP) on T2-weighted magnetic res-
onance imaging, which reflects the focal accumulation of iron in this area [119].
The substantia nigra (SN) is also affected in patients with CoPAN and to a lesser
extent in patients with PKAN. Iron accumulation correlates with neural damage and
mitochondrial lesions and is often associated with axonal expansions (spheroids) but
also appears as a granular form in perivascular sites [64].
Although iron accumulation is a hallmark of PKAN and CoPAN, its relation-
ship with CoA dysfunctional biosynthesis remains unclear. CoA is a key molecule
involved in more than 100 metabolic processes, among which CoA derivatives are
crucial substrates for ATP generation mediated by the tricarboxylic acid cycle, fatty
acid metabolism, cholesterol and ketone body biosynthesis, and histone and non-
histone protein acetylation [1]. The CoA biosynthetic pathway involves five universal
enzymatic steps that use pantothenate (vitamin B5), ATP, and cysteine. This pathway
is initiated by PANK2, which converts pantothenic acid into 4 -phosphopantothenic
162 S. Levi et al.
acid. The last two steps of CoA biosynthesis are mediated by COASY. Although
these processes are vital in any cell type, researchers have not yet explained why the
disease primarily affects the central nervous system.
Several animal models have been developed in an attempt to clarify the molecular
events that generate these disorders. Pank2 null mice showed reduced growth, male
infertility due to azoospermia [67], and impaired mitochondrial function [16], but did
not suffer from movement disorders or exhibit signs of neurodegeneration, implying
that the other Pank genes may compensate for the Pank2 deficiency in mice. Only by
modulating the fat content in the diet of these mice, it was possible to elicit neurode-
generation and cause a movement disorder associated with a general bioenergetic
failure [15]. The Drosophila Pank2 KO model (fumble) displays brain lesions and
defective neurological functions, resulting in severe motor impairment [113]. The
downregulation of pank2 in zebra fish induces defects in neuronal development and
in the formation of the vascular system [118].
A yeast model was used to study a human COASY pathogenic mutation. It showed
mitochondrial dysfunction, and both the iron and lipid contents were altered [12].
Downregulation of zebra fish coasy expression led to a generalized reduction in size,
poor definition of brain structures, and alteration of the vasculature [57]. Although
all of these animal models were informative tools for studying pathological mecha-
nisms, they share only a few neuropathological signs with milder severity than the
human disorder, limiting their impact for predicting novel therapies [71]. In fact,
while presenting some levels of neurodegeneration, these models lack any evidence
of brain iron mishandling, preventing researchers from obtaining any insights into
the causative link between CoA deficiency and brain iron deposition. Fibroblasts
from patients with PKAN have been instrumental in revealing some defects in mito-
chondrial activity and iron metabolism associated with the PANK2 deficiency [100];
however, their specific contributions to the pathological neurodegenerative processes
have not been identified in these cells.
Insights into the neurodegenerative process began to emerge in an investigation
of human neurons differentiated from induced pluripotent stem cells (iPSC) [87].
Compared with neurons from 3 healthy subjects, iPSC-derived glutamatergic neurons
from 3 patients with PKAN showed alterations in mitochondrial function and impair-
ments in mitochondrial iron-dependent biosynthesis that caused iron dyshomeostasis
and subsequently increased ROS production, leading to major membrane excitability
defects. In particular, the inability of the cells to maintain Fe/S biosynthesis simulates
a cellular iron-deficient phenotype that promotes cellular iron uptake and increases
the translation of TfR1 [87]. In addition, CoA supplementation prevented neuronal
death and ROS formation by restoring mitochondrial and neuronal functions, sug-
gesting that a CoA treatment is a possible therapeutic intervention [87]. A second
paper analyzed iPSC-derived cortical neuronal cells obtained from fibroblasts from
a patient with atypical PKAN [4]. Consistent with the milder phenotype, the Pank2
peptide was not abolished as described in the previous study but was present at lower
levels than in the control cells. In this case, CoA homeostasis and cellular iron han-
dling were normal; however, mitochondrial function was affected, with increased
generation of reactive oxygen species and lipid peroxidation in patient-derived neu-
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 163
rons [4]. Notably, an iron chelator treatment exacerbated the mitochondrial phenotype
in neuronal cells from both control subjects and patients. Thus, both of these inves-
tigations using human neurons concluded that mitochondrial impairment is an early
feature of the disease process and the subsequent ROS formation plays a main role in
the pathogenesis; however, the iron chelator treatment did not appear to be a viable
option. Indeed, the data on the efficacy of the DFP treatment collected from different
clinical trials are still under investigation. Two pilot studies confirmed the safety and
efficacy of the DFP treatment in reducing brain iron levels in patients with PKAN
[19, 119], but the results from the still ongoing clinical trial involving approximately
100 patients with PKAN are needed to define the clinical outcomes of iron chelator
treatments.
Since sequencing has become a method for diagnosing NBIA, many similar cases
in terms of symptomatology have been distinguished in different groups based on
the genes in which the mutations were identified. The main group is composed of
disorders associated with mutations in genes encoding proteins involved in lipid
homeostasis. These diseases typically begin in childhood with symptoms such as
developmental delay, regression or attention deficits associated with hyperactivity,
spastic paraparesis, dystonia, and visual pathway disorders. Atypical variants begin
in adolescence or adulthood, with a variable combination of dystonia, dysarthria,
parkinsonism, and psychiatric symptoms. Iron accumulation is not a universal feature
at the time when symptoms begin and is often documented only in the later stages,
suggesting that iron overload, which is typically present in the GP and sometimes in
the SN, is not the primary cause of the disease but contributes to neurodegeneration
[29].
The first pathology to be distinguished from PKAN by sequencing was PLAN (or
INAD, infantile neuroaxonal dystrophy, OMIM#256600), which is caused by defects
in the PLA2G6 gene located on chromosome 22q13.1 that encodes a calcium-
independent phospholipase involved in cell membrane homeostasis [116]. Defects in
the protein may lead to altered lipid composition of the plasma, vesicular, and endo-
somal membranes, with subsequent alterations in membrane maintenance, causing
neuronal damage [75]. This feature typically occurs in the first year of life as neu-
roaxonal childhood dystrophy with psychomotor regression and axial hypotonia that
164 S. Levi et al.
MPAM accounts for approximately 30% of the NBIA cases. It is caused by a mutation
in C19orf12 (OMIM*614297) that encodes a mitochondrial protein with an unknown
function but is probably involved in fatty acid metabolism and in the degradation of
the branched amino acid chains [46, 88]. The main symptoms of MPAN are pro-
gressive spastic para/tetraparesis, generalized dystonia, motor axonal neuropathy,
parkinsonism, psychiatric symptoms, retinal abnormalities, and optic atrophy [46,
50, 70]. Patients with MPAN develop Lewy bodies containing α-synuclein in the
GP and midbrain, as well as perivascular iron deposits similar to those observed in
patients with PKAN [49]. The C19orf12-encoded peptide is a transmembrane glycine
zipper-containing protein with two isoforms originating from different start codons.
In the literature, few reports have characterized the protein. An analysis of HeLa sub-
cellular fractions indicated that the longer isoform was located in the mitochondria,
endoplasmic reticulum, and a small fraction in the MAM [107], suggesting a role
in the transport of phospholipids. Fibroblasts from one patient showed an alteration
of mitochondrial calcium homeostasis and increased H2 O2 -induced apoptosis com-
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 165
pared to controls [107]. The only available animal disease model is the knockdown
of the C19orf12 orthologues in Drosophila that showed signs of neurological defects
and the presence of vacuoles in the brain and optical lobe, but not iron accumulation
[53].
Two other genes involved in lipid homeostasis have recently been identified as
causative for NBIA [28]. The genes are CRAT (OMIM*600184), which is involved
in the transfer of acyl groups from carnitine to CoA and in the transport of fatty acids
for beta-oxidation, and REPS1 (OMIM*614825), which is engaged in endocytosis
and vesicle transport. Interestingly, the authors also described a new perspective of
the NBIA pathogenic mechanism. They observed a common phenotype in fibrob-
lasts derived from patients with CRAT and REPS1 mutations, as well as fibroblasts
derived from patients with PANK2, PLA2G6, C19orf12, and FA2H mutations. All
these cells exhibited reduced TfR1 palmitoylation that affected the recycling of the
receptor [28]. This anomaly in cellular iron incorporation led to iron overload that
was decreased by treatment with the antimalarial drug artesunate. This drug promotes
receptor palmitoylation, restoring the normal TfR1 recycling [28]. This report was
the first time to experimentally confirm a common pathogenic mechanism for NBIA
disorders.
Although the role of lysosomes in iron metabolism is known [69], some lysosomal
disorders have only recently been included in the NBIA categories. These disorders
are caused by mutations in genes encoding lysosomal proteins and show a variable age
of onset and severity of the phenotype, depending on the degree of dysfunctionality
of the mutated proteins. An explanation for why only a portion of the affected patients
exhibit iron overload in the brain is unknown, and researchers have proposed that
iron is not the primary cause of the disease in this case [29].
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 167
A new candidate NBIA disorder was reported in a recent study that identified iron
deposition in the brains of 3 patients afflicted with an autosomal recessive form
of hereditary spastic paraplegia (HSP) (OMIM#602296) [98] that is characterized
by early-onset developmental delay, tetraparesis, motor function deterioration, and
intellectual disability. This very rare neurodevelopmental disorder is due to mutations
in AP4M1. The AP4M1 gene encodes the subunits of adaptor protein 4 (AP-4), a
heterotetrameric complex involved in intracellular protein trafficking [24]. No data
on iron involvement in the pathophysiology of the disorder have been reported;
however, the finding that fibroblasts from AP-4-deficient patients and neurons from
AP-4-deficient mice show impaired autophagy further emphasizes the stringent link
between alterations in autophagy and iron deposition.
Two other disorders are classified as NBIA, although not all patients show signs of
brain iron deposition. These disorders are briefly described here because published
data concerning the role of iron in the pathological process do not exist.
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 169
9.7 Conclusions
tion, where iron is required for its biosynthesis [97]. Membrane remodeling and
autophagy appear to be essential to avoid iron deposition in the brain. In particular,
the impaired ability of cells to disrupt the iron-containing macromolecules (i.e., fer-
ritin) and organelles (mitochondria) through the lysosomal pathway appears to be a
common phenotype in patients with various NBIAs.
Although additional in vitro and in vivo studies are needed to define whether
iron dysregulation has a primary or an auxiliary role in the pathophysiology of these
diseases, the clarification of the pathogenic mechanisms of NBIAs will add important
knowledge required for a better understanding of the relationship between iron and
the neurodegenerative process that is necessary to define the therapeutic options.
References
1. Akram M (2014) Citric acid cycle and role of its intermediates in metabolism. Cell Biochem
Biophys 68:475–478
2. Alazami AM, Al-Saif A, Al-Semari A, Bohlega S, Zlitni S, Alzahrani F, Bavi P, Kaya N,
Colak D, Khalak H, Baltus A, Peterlin B, Danda S, Bhatia KP, Schneider SA, Sakati N,
Walsh CA, Al-Mohanna F, Meyer B, Alkuraya FS (2008) Mutations in C2orf37, encoding a
nucleolar protein, cause hypogonadism, alopecia, diabetes mellitus, mental retardation, and
extrapyramidal syndrome. Am J Hum Genet 83:684–691
3. Annesi G, Gagliardi M, Iannello G, Quattrone A (2016) Mutational analysis of COASY in
an Italian patient with NBIA. Parkinsonism Relat Disord 28:150–151
4. Arber C, Angelova PR, Wiethoff S, Tsuchiya Y, Mazzacuva F, Preza E, Bhatia KP, Mills K,
Gout I, Abramov AY, Hardy J, Duce JA, Houlden H, Wray S (2017) iPSC-derived neuronal
models of PANK2-associated neurodegeneration reveal mitochondrial dysfunction contribut-
ing to early disease. PLoS ONE 12:e0184104
5. Baraibar MA, Barbeito AG, Muhoberac BB, Vidal R (2008) Iron-mediated aggregation and
a localized structural change characterize ferritin from a mutant light chain polypeptide that
causes neurodegeneration. J Biol Chem 283:31679–31689
6. Baraibar MA, Muhoberac BB, Garringer HJ, Hurley TD, Vidal R (2010) Unraveling of the
E-helices and disruption of 4-fold pores are associated with iron mishandling in a mutant
ferritin causing neurodegeneration. J Biol Chem 285:1950–1956
7. Barbeito AG, Garringer HJ, Baraibar MA, Gao X, Arredondo M, Nunez MT, Smith MA,
Ghetti B, Vidal R (2009) Abnormal iron metabolism and oxidative stress in mice expressing
a mutant form of the ferritin light polypeptide gene. J Neurochem 109:1067–1078
8. Barbeito AG, Levade T, Delisle MB, Ghetti B, Vidal R (2010) Abnormal iron metabolism in
fibroblasts from a patient with the neurodegenerative disease hereditary ferritinopathy. Mol
Neurodegener 5:50
9. Beck G, Shinzawa K, Hayakawa H, Baba K, Yasuda T, Sumi-Akamaru H, Tsujimoto Y,
Mochizuki H (2015) Deficiency of calcium-independent phospholipase A2 Beta induces
brain iron accumulation through upregulation of divalent metal transporter 1. PLoS ONE
10:e0141629
10. Beck G, Sugiura Y, Shinzawa K, Kato S, Setou M, Tsujimoto Y, Sakoda S, Sumi-Akamaru
H (2011) Neuroaxonal dystrophy in calcium-independent phospholipase A2beta deficiency
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 171
results from insufficient remodeling and degeneration of mitochondrial and presynaptic mem-
branes. J Neurosci 31:11411–11420
11. Behrens MI, Bruggemann N, Chana P, Venegas P, Kagi M, Parrao T, Orellana P, Garrido C,
Rojas CV, Hauke J, Hahnen E, Gonzalez R, Seleme N, Fernandez V, Schmidt A, Binkofski
F, Kompf D, Kubisch C, Hagenah J, Klein C, Ramirez A (2010) Clinical spectrum of Kufor-
Rakeb syndrome in the Chilean kindred with ATP13A2 mutations. Mov Disord 25:1929–1937
12. Berti CC, Dallabona C, Lazzaretti M, Dusi S, Tosi E, Tiranti V, Goffrini P (2015) Modeling
human Coenzyme A synthase mutation in yeast reveals altered mitochondrial function, lipid
content and iron metabolism. Microb Cell 2:126–135
13. Bertoli-Avella AM, Garcia-Aznar JM, Brandau O, Al-Hakami F, Yuksel Z, Marais A, Gruning
NM, Abbasi Moheb L, Paknia O, Alshaikh N, Alameer S, Marafi MJ, Al-Mulla F, Al-Sannaa
N, Rolfs A, Bauer P (2018) Biallelic inactivating variants in the GTPBP2 gene cause a
neurodevelopmental disorder with severe intellectual disability. Eur J Hum Genet 26:592–598
14. Brissot P, Ropert M, Le Lan C, Loreal O (2012) Non-transferrin bound iron: a key role in iron
overload and iron toxicity. Biochim Biophys Acta 1820:403–410
15. Brunetti D, Dusi S, Giordano C, Lamperti C, Morbin M, Fugnanesi V, Marchet S, Fagiolari G,
Sibon O, Moggio M, d’Amati G, Tiranti V (2014) Pantethine treatment is effective in recov-
ering the disease phenotype induced by ketogenic diet in a pantothenate kinase-associated
neurodegeneration mouse model. Brain 137:57–68
16. Brunetti D, Dusi S, Morbin M, Uggetti A, Moda F, D’Amato I, Giordano C, d’Amati G, Cozzi
A, Levi S, Hayflick S, Tiranti V (2012) Pantothenate kinase-associated neurodegeneration:
altered mitochondria membrane potential and defective respiration in Pank2 knock-out mouse
model. Hum Mol Genet 21:5294–5305
17. Capoccia S, Maccarinelli F, Buffoli B, Rodella LF, Cremona O, Arosio P, Cirulli F (2015)
Behavioral characterization of mouse models of neuroferritinopathy. PLoS ONE 10:e0118990
18. Chinnery PF, Crompton DE, Birchall D, Jackson MJ, Coulthard A, Lombes A, Quinn N,
Wills A, Fletcher N, Mottershead JP, Cooper P, Kellett M, Bates D, Burn J (2007) Clinical
features and natural history of neuroferritinopathy caused by the FTL1 460InsA mutation.
Brain 130:110–119
19. Cossu G, Abbruzzese G, Matta G, Murgia D, Melis M, Ricchi V, Galanello R, Barella S,
Origa R, Balocco M, Pelosin E, Marchese R, Ruffinengo U, Forni GL (2014) Efficacy and
safety of deferiprone for the treatment of pantothenate kinase-associated neurodegeneration
(PKAN) and neurodegeneration with brain iron accumulation (NBIA): results from a four
years follow-up. Parkinsonism Relat Disord 20:651–654
20. Cozzi A, Rovelli E, Frizzale G, Campanella A, Amendola M, Arosio P, Levi S (2010) Oxida-
tive stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy.
Neurobiol Dis 37:77–85
21. Cozzi A, Santambrogio P, Corsi B, Campanella A, Arosio P, Levi S (2006) Characterization
of the l-ferritin variant 460InsA responsible of a hereditary ferritinopathy disorder. Neurobiol
Dis 23:644–652
22. Curtis AR, Fey C, Morris CM, Bindoff LA, Ince PG, Chinnery PF, Coulthard A, Jackson MJ,
Jackson AP, McHale DP, Hay D, Barker WA, Markham AF, Bates D, Curtis A, Burn J (2001)
Mutation in the gene encoding ferritin light polypeptide causes dominant adult-onset basal
ganglia disease. Nat Genet 28:350–354
23. de Figueiredo P, Doody A, Polizotto RS, Drecktrah D, Wood S, Banta M, Strang MS, Brown
WJ (2001) Inhibition of transferrin recycling and endosome tubulation by phospholipase A2
antagonists. J Biol Chem 276:47361–47370
24. De Pace R, Skirzewski M, Damme M, Mattera R, Mercurio J, Foster AM, Cuitino L, Jarnik
M, Hoffmann V, Morris HD, Han TU, Mancini GMS, Buonanno A, Bonifacino JS (2018)
Altered distribution of ATG9A and accumulation of axonal aggregates in neurons from a
mouse model of AP-4 deficiency syndrome. PLoS Genet 14:e1007363
25. de Tezanos Pinto F, Adamo HP (2018) The strategic function of the P5-ATPase ATP13A2 in
toxic waste disposal. Neurochem Int 112:108–113
172 S. Levi et al.
26. Devos D, Tchofo PJ, Vuillaume I, Destee A, Batey S, Burn J, Chinnery PF (2009) Clinical
features and natural history of neuroferritinopathy caused by the 458dupA FTL mutation.
Brain 132:e109
27. di Patti MC, Maio N, Rizzo G, De Francesco G, Persichini T, Colasanti M, Polticelli F,
Musci G (2009) Dominant mutants of ceruloplasmin impair the copper loading machinery in
aceruloplasminemia. J Biol Chem 284:4545–4554
28. Drecourt A, Babdor J, Dussiot M, Petit F, Goudin N, Garfa-Traore M, Habarou F, Bole-Feysot
C, Nitschke P, Ottolenghi C, Metodiev MD, Serre V, Desguerre I, Boddaert N, Hermine O,
Munnich A, Rotig A (2018) Impaired transferrin receptor palmitoylation and recycling in
neurodegeneration with brain iron accumulation. Am J Hum Genet 102:266–277
29. Dusek P, Schneider SA (2012) Neurodegeneration with brain iron accumulation. Curr Opin
Neurol 25:499–506
30. Dusek P, Schneider SA (2013) Neurodegenerative disorder with brain iron accumulation pre-
viously known as SENDA syndrome now genetically determined. Mov Disord 28:1051–1052
31. Dusi S, Valletta L, Haack TB, Tsuchiya Y, Venco P, Pasqualato S, Goffrini P, Tigano M,
Demchenko N, Wieland T, Schwarzmayr T, Strom TM, Invernizzi F, Garavaglia B, Gregory
A, Sanford L, Hamada J, Bettencourt C, Houlden H, Chiapparini L, Zorzi G, Kurian MA,
Nardocci N, Prokisch H, Hayflick S, Gout I, Tiranti V (2014) Exome sequence reveals muta-
tions in CoA synthase as a cause of neurodegeneration with brain iron accumulation. Am J
Hum Genet 94:11–22
32. Eiberg H, Hansen L, Korbo L, Nielsen IM, Svenstrup K, Bech S, Pinborg LH, Friberg L,
Hjermind LE, Olsen OR, Nielsen JE (2012) Novel mutation in ATP13A2 widens the spectrum
of Kufor-Rakeb syndrome (PARK9). Clin Genet 82:256–263
33. Evers C, Seitz A, Assmann B, Opladen T, Karch S, Hinderhofer K, Granzow M, Paramasivam
N, Eils R, Diessl N, Bartram CR, Moog U (2017) Diagnosis of CoPAN by whole exome
sequencing: waking up a sleeping tiger’s eye. Am J Med Genet A
34. Ferdinandusse S, Kostopoulos P, Denis S, Rusch H, Overmars H, Dillmann U, Reith W, Haas
D, Wanders RJ, Duran M, Marziniak M (2006) Mutations in the gene encoding peroxisomal
sterol carrier protein X (SCPx) cause leukencephalopathy with dystonia and motor neuropathy.
Am J Hum Genet 78:1046–1052
35. Finkenstedt A, Wolf E, Hofner E, Gasser BI, Bosch S, Bakry R, Creus M, Kremser C, Schocke
M, Theurl M, Moser P, Schranz M, Bonn G, Poewe W, Vogel W, Janecke AR, Zoller H (2010)
Hepatic but not brain iron is rapidly chelated by deferasirox in aceruloplasminemia due to a
novel gene mutation. J Hepatol 53:1101–1107
36. Garringer HJ, Irimia JM, Li W, Goodwin CB, Richine B, Acton A, Chan RJ, Peacock M,
Muhoberac BB, Ghetti B, Vidal R (2016) Effect of systemic iron overload and a chelation
therapy in a mouse model of the neurodegenerative disease hereditary Ferritinopathy. PLoS
ONE 11:e0161341
37. Gillis WQ, Kirmizitas A, Iwasaki Y, Ki DH, Wyrick JM, Thomsen GH (2016) Gtpbp2 is a
positive regulator of Wnt signaling and maintains low levels of the Wnt negative regulator
Axin. Cell Commun Signal 14:15
38. Gonzalez-Cuyar LF, Perry G, Miyajima H, Atwood CS, Riveros-Angel M, Lyons PF, Siedlak
SL, Smith MA, Castellani RJ (2008) Redox active iron accumulation in aceruloplasminemia.
Neuropathology 28:466–471
39. Gregory A, Hayflick SJ (2011) Genetics of neurodegeneration with brain iron accumulation.
Curr Neurol Neurosci Rep 11:254–261
40. Gregory A, Venkateswaran S, Hayflick SJ (2011) Fatty acid hydroxylase-associated neurode-
generation. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K,
Amemiya A (eds) GeneReviews((R)). Seattle, pp 1993–2018
41. Haack TB, Hogarth P, Kruer MC, Gregory A, Wieland T, Schwarzmayr T, Graf E, Sanford
L, Meyer E, Kara E, Cuno SM, Harik SI, Dandu VH, Nardocci N, Zorzi G, Dunaway T,
Tarnopolsky M, Skinner S, Frucht S, Hanspal E, Schrander-Stumpel C, Heron D, Mignot
C, Garavaglia B, Bhatia K, Hardy J, Strom TM, Boddaert N, Houlden HH, Kurian MA,
Meitinger T, Prokisch H, Hayflick SJ (2012) Exome sequencing reveals de novo WDR45
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 173
58. Kinghorn KJ, Castillo-Quan JI, Bartolome F, Angelova PR, Li L, Pope S, Cocheme HM,
Khan S, Asghari S, Bhatia KP, Hardy J, Abramov AY, Partridge L (2015) Loss of PLA2G6
leads to elevated mitochondrial lipid peroxidation and mitochondrial dysfunction. Brain
138:1801–1816
59. Kono S (2012) Aceruloplasminemia. Curr Drug Targets 13:1190–1199
60. Kono S (2013) Aceruloplasminemia: an update. Int Rev Neurobiol 110:125–151
61. Kono S, Miyajima H (2006) Molecular and pathological basis of aceruloplasminemia. Biol
Res 39:15–23
62. Kono S, Suzuki H, Oda T, Shirakawa K, Takahashi Y, Kitagawa M, Miyajima H (2007)
Cys-881 is essential for the trafficking and secretion of truncated mutant ceruloplasmin in
aceruloplasminemia. J Hepatol 47:844–850
63. Kono S, Yoshida K, Tomosugi N, Terada T, Hamaya Y, Kanaoka S, Miyajima H (2010) Bio-
logical effects of mutant ceruloplasmin on hepcidin-mediated internalization of ferroportin.
Biochim Biophys Acta 1802:968–975
64. Kruer MC, Hiken M, Gregory A, Malandrini A, Clark D, Hogarth P, Grafe M, Hayflick SJ,
Woltjer RL (2011) Novel histopathologic findings in molecularly-confirmed pantothenate
kinase-associated neurodegeneration. Brain 134:947–958
65. Kruer MC, Paisan-Ruiz C, Boddaert N, Yoon MY, Hama H, Gregory A, Malandrini A, Wolt-
jer RL, Munnich A, Gobin S, Polster BJ, Palmeri S, Edvardson S, Hardy J, Houlden H,
Hayflick SJ (2010) Defective FA2H leads to a novel form of neurodegeneration with brain
iron accumulation (NBIA). Ann Neurol 68:611–618
66. Kubota A, Hida A, Ichikawa Y, Momose Y, Goto J, Igeta Y, Hashida H, Yoshida K, Ikeda S,
Kanazawa I, Tsuji S (2009) A novel ferritin light chain gene mutation in a Japanese family with
neuroferritinopathy: description of clinical features and implications for genotype-phenotype
correlations. Mov Disord 24:441–445
67. Kuo YM, Duncan JL, Westaway SK, Yang H, Nune G, Xu EY, Hayflick SJ, Gitschier J
(2005) Deficiency of pantothenate kinase 2 (Pank2) in mice leads to retinal degeneration and
azoospermia. Hum Mol Genet 14:49–57
68. Kurian MA, Hayflick SJ (2013) Pantothenate kinase-associated neurodegeneration (PKAN)
and PLA2G6-associated neurodegeneration (PLAN): review of two major neurodegeneration
with brain iron accumulation (NBIA) phenotypes. Int Rev Neurobiol 110:49–71
69. Kurz T, Eaton JW, Brunk UT (2011) The role of lysosomes in iron metabolism and recycling.
Int J Biochem Cell Biol 43:1686–1697
70. Langwinska-Wosko E, Skowronska M, Kmiec T, Czlonkowska A (2016) Retinal and optic
nerve abnormalities in neurodegeneration associated with mutations in C19orf12 (MPAN). J
Neurol Sci 370:237–240
71. Levi S, Finazzi D (2014) Neurodegeneration with brain iron accumulation: update on
pathogenic mechanisms. Front Pharmacol 5:99
72. Levi S, Rovida E (2015) Neuroferritinopathy: from ferritin structure modification to patho-
genetic mechanism. Neurobiol Dis 81:134–143
73. Maccarinelli F, Pagani A, Cozzi A, Codazzi F, Di Giacomo G, Capoccia S, Rapino S, Finazzi
D, Politi LS, Cirulli F, Giorgio M, Cremona O, Grohovaz F, Levi S (2015) A novel neu-
roferritinopathy mouse model (FTL 498InsTC) shows progressive brain iron dysregulation,
morphological signs of early neurodegeneration and motor coordination deficits. Neurobiol
Dis 81:119–133
74. Maciel P, Cruz VT, Constante M, Iniesta I, Costa MC, Gallati S, Sousa N, Sequeiros J,
Coutinho P, Santos MM (2005) Neuroferritinopathy: missense mutation in FTL causing early-
onset bilateral pallidal involvement. Neurology 65:603–605
75. Malik I, Turk J, Mancuso DJ, Montier L, Wohltmann M, Wozniak DF, Schmidt RE, Gross RW,
Kotzbauer PT (2008) Disrupted membrane homeostasis and accumulation of ubiquitinated
proteins in a mouse model of infantile neuroaxonal dystrophy caused by PLA2G6 mutations.
Am J Pathol 172:406–416
76. Mancuso M, Davidzon G, Kurlan RM, Tawil R, Bonilla E, Di Mauro S, Powers JM (2005)
Hereditary ferritinopathy: a novel mutation, its cellular pathology, and pathogenetic insights.
J Neuropathol Exp Neurol 64:280–294
9 Iron Pathophysiology in Neurodegeneration with Brain Iron … 175
77. Mari F, Berti B, Romano A, Baldacci J, Rizzi R, Grazia Alessandri M, Tessa A, Procopio
E, Rubegni A, Lourenco CM, Simonati A, Guerrini R, Santorelli FM (2018) Clinical and
neuroimaging features of autosomal recessive spastic paraplegia 35 (SPG35): case reports,
new mutations, and brief literature review. Neurogenetics 19:123–130
78. McNeill A, Birchall D, Hayflick SJ, Gregory A, Schenk JF, Zimmerman EA, Shang H, Miya-
jima H, Chinnery PF (2008) T2* and FSE MRI distinguishes four subtypes of neurodegener-
ation with brain iron accumulation. Neurology 70:1614–1619
79. McNeill A, Chinnery PF (2011) Neurodegeneration with brain iron accumulation. Handb Clin
Neurol 100:161–172
80. Miyajima H (2003) Aceruloplasminemia, an iron metabolic disorder. Neuropathology
23:345–350
81. Miyajima H (2015) Aceruloplasminemia. Neuropathology 35:83–90
82. Miyajima H, Nishimura Y, Mizoguchi K, Sakamoto M, Shimizu T, Honda N (1987) Familial
apoceruloplasmin deficiency associated with blepharospasm and retinal degeneration. Neu-
rology 37:761–767
83. Moutton S, Fergelot P, Trocello JM, Plante-Bordeneuve V, Houcinat N, Wenisch E, Larue
V, Brugieres P, Clot F, Lacombe D, Arveiler B, Goizet C (2014) A novel FTL mutation
responsible for neuroferritinopathy with asymmetric clinical features and brain anomalies.
Parkinsonism Relat Disord 20:935–937
84. Nishida K, Garringer HJ, Futamura N, Funakawa I, Jinnai K, Vidal R, Takao M (2014) A novel
ferritin light chain mutation in neuroferritinopathy with an atypical presentation. J Neurol Sci
342:173–177
85. Ogimoto M, Anzai K, Takenoshita H, Kogawa K, Akehi Y, Yoshida R, Nakano M,
Yoshida K, Ono J (2011) Criteria for early identification of aceruloplasminemia. Intern Med
50:1415–1418
86. Ohta E, Nagasaka T, Shindo K, Toma S, Nagasaka K, Ohta K, Shiozawa Z (2008) Neurofer-
ritinopathy in a Japanese family with a duplication in the ferritin light chain gene. Neurology
70:1493–1494
87. Orellana DI, Santambrogio P, Rubio A, Yekhlef L, Cancellieri C, Dusi S, Giannelli SG, Venco
P, Mazzara PG, Cozzi A, Ferrari M, Garavaglia B, Taverna S, Tiranti V, Broccoli V, Levi S
(2016) Coenzyme A corrects pathological defects in human neurons of PANK2-associated
neurodegeneration. EMBO Mol Med 8:1197–1211
88. Panteghini C, Zorzi G, Venco P, Dusi S, Reale C, Brunetti D, Chiapparini L, Zibordi F, Siegel
B, Garavaglia B, Simonati A, Bertini E, Nardocci N, Tiranti V (2012) C19orf12 and FA2H
mutations are rare in Italian patients with neurodegeneration with brain iron accumulation.
Semin Pediatr Neurol 19:75–81
89. Patel BN, Dunn RJ, Jeong SY, Zhu Q, Julien JP, David S (2002) Ceruloplasmin regulates iron
levels in the CNS and prevents free radical injury. J Neurosci 22:6578–6586
90. Potter KA, Kern MJ, Fullbright G, Bielawski J, Scherer SS, Yum SW, Li JJ, Cheng H, Han
X, Venkata JK, Khan PA, Rohrer B, Hama H (2011) Central nervous system dysfunction in
a mouse model of FA2H deficiency. Glia 59:1009–1021
91. Powers JM (2006) p53-mediated apoptosis, neuroglobin overexpression, and globin deposits
in a patient with hereditary ferritinopathy. J Neuropathol Exp Neurol 65:716–721
92. Proikas-Cezanne T, Waddell S, Gaugel A, Frickey T, Lupas A, Nordheim A (2004) WIPI-
1alpha (WIPI49), a member of the novel 7-bladed WIPI protein family, is aberrantly expressed
in human cancer and is linked to starvation-induced autophagy. Oncogene 23:9314–9325
93. Rajagopalan S, Rane A, Chinta SJ, Andersen JK (2016) Regulation of ATP13A2 via PHD2-
HIF1alpha signaling is critical for cellular iron homeostasis: implications for Parkinson’s
disease. J Neurosci 36:1086–1095
94. Ramirez A, Heimbach A, Grundemann J, Stiller B, Hampshire D, Cid LP, Goebel I, Mubaidin
AF, Wriekat AL, Roeper J, Al-Din A, Hillmer AM, Karsak M, Liss B, Woods CG, Behrens
MI, Kubisch C (2006) Hereditary parkinsonism with dementia is caused by mutations in
ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet 38:1184–1191
176 S. Levi et al.
Abstract Iron has been proposed to be responsible for neuronal loss in several
diseases of the central nervous system, including Alzheimer’s disease (AD), Parkin-
son’s disease (PD), stroke, Friedreich’s ataxia (FRDA), multiple sclerosis (MS),
amyotrophic lateral sclerosis (ALS). In many diseases, abnormal accumulation of
brain iron in disease-affected area has been observed, without clear knowledge of the
contribution of iron overload to pathogenesis. Recent evidences implicate that key
proteins involved in the disease pathogenesis may also participate in cellular iron
metabolism, suggesting that the imbalance of brain iron homeostasis is associated
with the diseases. Considering the complicated regulation of iron homeostasis within
the brain, a thorough understanding of the molecular events leading to this phenotype
is still to be investigated. However, current understanding has already provided the
basis for the diagnosis and treatment of iron-related CNS diseases, which will be
reviewed here.
10.1 Introduction
In the central nervous system (CNS), iron is involved in multiple crucial biological
processes including oxygen transportation, DNA synthesis, mitochondrial respira-
tion, myelin synthesis, and the synthesis and metabolism of neurotransmitters [1].
It plays an important role in electron transfer and is cofactors for varies enzymes
[2]. Iron homoeostasis is needed to maintain normal physiological brain function,
whereas abnormal iron homoeostasis can induce cellular damage through reactive
oxygen species (ROS), notably the hydroxyl radical [3]. ROS can cause the oxidation
and modification of lipids, proteins, carbohydrates, and DNA [4, 5]. Peroxidation of
polyunsaturated fatty acids in membrane lipids by ROS can generate highly reac-
tive aldehydes, such as 4-hydroxynonenal, which irreversibly modify proteins by
carbonylation [6–8]. ROS can induce the release of iron from mitochondrial iron-
sulfur cluster proteins of the respiratory chain and other iron storage proteins, which
will lead to further ROS production via Fenton’s reaction [9]. Iron-related CNS dis-
eases that result from iron toxicity can lead to apoptosis [10] and ferroptosis [11],
an iron-specific form of non-apoptotic cell death [12].
In various iron-related CNS diseases, changes in iron homoeostasis result in
altered cellular iron distribution and accumulation [13–21]. Magnetic resonance
imaging (MRI) can often identify these changes, thus providing a potential diag-
nostic biomarker of iron-related CNS diseases. An important avenue to reduce iron
accumulation is the use of iron chelators that are able to cross the blood–brain barrier,
penetrate cells, and reduce excessive iron accumulation, thereby affording neuropro-
tection. These will provide new ideas for the diagnostics and treatment of iron-related
CNS diseases.
impairment [26]. These data indicated that iron in specific brain regions may be used
as a biomarker to evaluate the progression of AD. Australian researchers have applied
quantitative susceptibility mapping (QSM), a relatively new MRI method sensitive
to tissue iron, to assess the relationship between iron, Aβ load, and cognitive decline.
They found that QSM level is associated with Aβ deposition, which is consistent
with brain iron increasing during the natural history of AD [27]. This study showed
that higher levels of brain iron can also predict clinical deterioration.
Ferritin is the major iron storage protein of the body; by using cerebrospinal fluid
(CSF) levels of ferritin as an index, researchers explored whether brain iron status
impacts the longitudinal outcomes in the AD Neuroimaging Initiative (ADNI) cohort.
Results showed that CSF ferritin levels were negatively associated with cognitive
performance over 7 year’s period and predicted the conversion from mild cognitive
impairment (MCI) to AD [28]. These findings reveal that ferritin levels in the CSF
may predict AD outcomes.
Some studies in postmortem PD brains also get the similar conclusion that iron
is overloaded in SN in PD. For instance, detection the concentration of metal ion in
SN with Electrothermal Atomic Absorption Spectrometry (ETAAS), discovered that
the labile iron concentration but not copper was significantly higher in PD patients
than in healthy control [37]. Lei et al. also found that iron elevation in PD patients
and demonstrated that it may be associated with tau deficiency [15].
Diseases that affect iron status also have influences on PD. Mutation of gene
encoding ferritin light polypeptide would cause iron deposition and lead to adult-
onset basal ganglia disease such as parkinsonism symptom [38, 39]. Ceruloplasmin
(Cp) is a copper oxidase and plays a role in iron export from cell [40]. About 176
PD patients from UK Brain Bank with increased SN iron level were examined for
mutations of Cp gene and found that five missense variations about Cp were related to
PD [41]. Mutation of pantothenate kinase 2 (PANK2) will lead to iron accumulation
in basal ganglia [42], while two siblings with this PANK2 mutation come from
consanguineous parents have developed parkinsonism [43].
10.2.3 Stroke
RLS is a chronic neurological disorder with unique clinical symptoms. The diag-
nosis of RLS used to be based on the subjective description of symptoms, lacking
of available biomarkers or objective tests [61]. Since RLS can be exacerbated by
systemic iron deficiency, all newly diagnosed patients as well as those with wors-
ening symptoms should have iron-level tests [61]. Clinical study showed that the
SN iron index, determined by MRI, significantly decreased in the early-onset RLS
patients. And there was a significant negative correlation between SN iron index and
the Johns Hopkins RLS severity scale [62]. In addition, iron deficiency was found
in red nucleus, pallidum, thalamus, and putamen of RLS patients [63, 64]. Further-
more, compared to healthy control, the CSF ferritin and transferrin levels from RLS
patients significantly decreased and increased, respectively [65].
The diagnosis of MS needs the integration of clinical, imaging, and laboratory evi-
dences. Besides, the magnetic field correlations value, indicating iron deposition, was
increased in the putamen, thalamus, and globus pallidus in MS patients compared
with the healthy control [66]. And MS lesions had 47 μg of iron per gram of tissue
more than the surrounding tissue on average, quantified by susceptibility-weighted
imaging [67]. In chronic MS, iron levels reduced in normal-appearing white matter
and correlated with disease duration. The ferritin and frataxin levels were upregulated
184 H. Xiong et al.
and downregulated in initial lesions, respectively [68]. Previous studies indicated that
the level of iron deposition might be related to disease duration and severity [69, 70].
Aging is the single highest risk factor for AD with an incidence of 50% in people over
the age of 85 [80]. Over more than thirty years of research, no disease-modifying
treatment has been approved by either the US Food and Drug Administration (FDA)
or the European Medicines Agency (EMA) for AD treatment.
Given the pathological iron accumulation in AD, iron removal from specific brain
region without causing a systematic iron deficiency should be taken into considera-
tion [81, 82]. McLachlan and colleagues [83] conducted a two-year, single-blinded
study to investigate whether the progression of dementia could be slowed by an
iron chelator, desferrioxamine (DFO), and showed a substantial reduction in the
rate of deterioration of daily living skills in 48 patients with Alzheimer’s disease
who were given DFO (125 mg intramuscularly twice daily, 5 days per week, for
24 months) when compared with Alzheimer’s disease given placebo. Despite the
promising results, no other clinical trials using DFO for AD have been investigated
so far. And yet, iron chelation using intranasal deferoxamine has been investigated
in mouse models of AD, was shown to reverse iron-induced memory deficits, and
inhibits amyloidogenic APP processing and Aβ aggregation [84–86].
5-Chloro-7-iodoquinolin-8-ol (clioquinol) is a moderate chelator of iron, copper,
and zinc, and for this reason, it was explored as a drug candidate for AD [87]. Indeed,
clioquinol has shown efficacy in patients with AD and models of diseases. Clioquinol
10 Diagnostics and Treatments of Iron-Related CNS Diseases 185
The primary treatment strategies for Parkinson’s disease include medications such as
carbidopa-levodopa, dopamine agonists, MAO-B inhibitors, as well as anticholiner-
gics, and deep brain stimulation surgery if necessary. Given the substantial evidences
linking iron accumulation with PD, a few treatments targeting iron were trialed.
The iron chelator deferiprone had been trialed for PD. A patient with symptoms
of dysarthria, orofacial dystonia, moderate parkinsonism, iron accumulation in the
substantia nigra, red nuclei, internal globi pallidi, and dentate nuclei detected by
T2*-weighted brain MRI had developed worsen during 5 years even was given med-
ications of levodopa, baclofen, oxitriptan, and buspirone. She was given 30 mg/kg
deferiprone per day for 32 months and found that her symptoms were improved 30%
after 6 months treatment, and the UPDRS decreased 30% after 1 year, while iron
accumulation in bilateral nuclei, substantia nigra were decreased, but not changed in
the red nuclei after 32 months [99]. PD is associated with iron deposition in SN and
iron deposition will increase oxidative damage. Grolez et al. carried out a pilot clini-
cal trial about treating PD patients with deferiprone or placebo for 6–12 months and
found that the iron level in SN and the UPDSR was significantly decreased; in addi-
tion, if the patients have gene mutation about iron metabolism, the effect of reducing
iron was stronger [100]. Devos et al. detected cytoprotection of deferiprone treated
cell models of PD or MPTP-inject mouse model of PD, and found a high level
186 H. Xiong et al.
10.3.3 Stroke
Injection of tissue plasminogen activator (tPA, also called alteplase) is the only
evidence-based treatment option for ischemic stroke [109]. However, limited by
the narrow therapeutic window (less than 4.5 h after the onset of ischemic stroke)
and other safety concerns such as potential bleeding in the brain, less than 5% of
patients have received this treatment [110]. Therefore, other effective therapies are
needed. Deferoxamine mesylate has exhibited neuroprotective effects by suppression
of iron-induced hydroxyl radical formation. In a clinical trial, the serum levels of per-
oxides had decreased and the levels of total radical-trapping antioxidant capacity had
increased after 3-day treatment with deferoxamine mesylate in stroke patients [111].
Tirilazad mesylate, a nonglucocorticoid, 21-aminosteroid, and iron-dependent per-
oxidation inhibitor, has been investigated. In a randomized, double-blinded, placebo-
controlled trial, patients with acute stroke were treated with the tirilazad mesylate
beginning at a median of 4.3 h after stroke. While the overall functional outcome
had not been improved measured by the Glasgow Outcome Scale and the Barthel
Index 3 months later [112], a higher dose showed 14% absolute reduction in mortal-
ity [113]. In a rat model of ischemic stroke, intranasally administered deferoxamine
improved the outcome [114], and inhibitors of ferroptosis protected from ischemic
injury in MCAO mouse model [115].
10 Diagnostics and Treatments of Iron-Related CNS Diseases 187
Since excess mitochondrial iron may worse the symptom of FRDA, iron chelators
were considered to treat FRDA. Deferoxamine cannot cross the blood–brain barrier
[117] and therefore might induce extracellular iron deficiency before the reduc-
tion of mitochondrial iron [118]. Another orally iron chelator deferiprone can cross
the blood–brain barrier with low affinity to iron [117]. Clinical trials indicated that
deferiprone at low concentrations had beneficial effects, but worse the symptoms of
FRDA at high doses [55, 119]. In another clinical trial, deferiprone and idebenone,
latter of which was an analog of coenzyme Q10 , were combined to treat FRDA. After
11 months of therapy, iron deposits in dentate nucleus, determined by MRI, as well as
heart hypertrophy parameters, were improved significantly. However, posture scores
and gait got worse [120].
Clinical studies suggested that in certain cases correcting the iron deficiency by
iron supplementation alone can relieve RLS without any further intervention [61].
In an open-label study, a single 1000 mg intravenous (IV) infusion of iron dextran
to treat RLS was evaluated. The results indicated that IV iron dextran significantly
improved the RLS symptom severity, duration, and total sleep time two weeks later.
In addition, brain iron concentration as determined by MRI was increased in the
SN and the prefrontal cortex [121]. In a randomized, parallel-group double-blinded
study, IV iron sucrose treatment significantly increased the CSF ferritin level and
reduced the RLS symptom severity, but did not change MRI iron index or periodic
leg movements of sleep [122].
Although iron chelation therapy works in animal model of MS [123], clinical studies
have failed to show its efficacy. In one clinical study, desferrioxamine was given to
9 patients up to 8 courses over 2 years. During the trial, 1 patient was improved, 3
patients unchanged, and 5 patients got worse, although the drug was well tolerated
[124, 125]. A preliminary data indicated that the symptoms of 2–4 patients with
progressive MS could be improved by the combination of iron depletion and ery-
188 H. Xiong et al.
thropoietin [126]. Therefore, the effectiveness of iron chelators for MS needs further
investigation.
Recently, iron chelation for ALS was assessed in a pilot clinical trial. The results
showed that the body mass index and the ALS Functional Rating Scale significantly
decreased for the first 3 months of deferiprone therapy. The iron levels in the cervical
spinal cord, motor cortex, and medulla oblongata reduced after deferiprone treatment.
These data encourage further attempt to test iron chelation for ALS.
10.4 Conclusion
Evidences have shown that iron deposition in brain was closely related with the
pathogenesis of multiple CNS diseases, and treatments to reduce iron in brain have
been investigated in clinical and animal studies. With positive outcomes, there are
still challenges to use iron chelation as therapy for brain disorders. Therefore, future
investigations are required to develop iron-trapping agents that are highly selective,
low affinity, BBB permeable, low in toxicity. With the right agent, the right time
window, and the right patients to intervene, iron will be a tractable target for CNS
diseases.
References
1. Crichton RR, Boelaert JR (2001) Inorganic biochemistry of iron metabolism: from molecular
mechanisms to clinical consequences. 2nd edn. Wiley
2. Rouault TA (2013) Iron metabolism in the CNS: implications for neurodegenerative diseases.
Nat Rev Neurosci 14:551–564. https://doi.org/10.1038/nrn3453
3. Crichton RR, Ward RJ (2008) Metal-based neurodegeneration: from molecular mechanisms
to therapeutic strategies. 2nd edn
4. Hentze MW, Muckenthaler MU, Galy B, Camaschella C (2010) Two to tango: regulation of
mammalian iron metabolism. Cell 142:24–38. https://doi.org/10.1016/j.cell.2010.06.028
5. Wang J, Pantopoulos K (2011) Regulation of cellular iron metabolism. Biochem J
434:365–381. https://doi.org/10.1042/BJ20101825
6. Catala A (2009) Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals
and oxidized phospholipids active in physiological and/or pathological conditions. Chem Phys
Lipids 157:1–11. https://doi.org/10.1016/j.chemphyslip.2008.09.004
7. Dalle-Donne I, Giustarini D, Colombo R, Rossi R, Milzani A (2003) Protein carbonylation
in human diseases. Trends Mol Med 9:169–176
8. Perluigi M, Coccia R, Butterfield DA (2012) 4-Hydroxy-2-nonenal, a reactive product of
lipid peroxidation, and neurodegenerative diseases: a toxic combination illuminated by redox
10 Diagnostics and Treatments of Iron-Related CNS Diseases 189
30. Powers KM et al (2009) Dietary fats, cholesterol and iron as risk factors for Parkinson’s
disease. Parkinsonism Relat Disord 15:47–52. https://doi.org/10.1016/j.parkreldis.2008.03.
002
31. Pichler I et al (2013) Serum iron levels and the risk of Parkinson disease: a Mendelian ran-
domization study. PLoS Med 10:e1001462. https://doi.org/10.1371/journal.pmed.1001462
32. Jimenez-Jimenez FJ et al (1998) Cerebrospinal fluid levels of transition metals in patients
with Parkinson’s disease. J Neural Transm (Vienna) 105:497–505. https://doi.org/10.1007/
s007020050073
33. Zecca L et al (2005) In vivo detection of iron and neuromelanin by transcranial sonography:
a new approach for early detection of substantia nigra damage. Mov Disord 20:1278–1285.
https://doi.org/10.1002/mds.20550
34. Becker G, Seufert J, Bogdahn U, Reichmann H, Reiners K (1995) Degeneration of sub-
stantia nigra in chronic Parkinson’s disease visualized by transcranial color-coded real-time
sonography. Neurology 45:182–184
35. Prestel J, Schweitzer KJ, Hofer A, Gasser T, Berg D (2006) Predictive value of transcranial
sonography in the diagnosis of Parkinson’s disease. Mov Disord 21:1763–1765. https://doi.
org/10.1002/mds.21054
36. Zhang J et al (2010) Characterizing iron deposition in Parkinson’s disease using susceptibility-
weighted imaging: an in vivo MR study. Brain Res 1330:124–130. https://doi.org/10.1016/j.
brainres.2010.03.036
37. Wypijewska A et al (2010) Iron and reactive oxygen species activity in parkinsonian substantia
nigra. Parkinsonism Relat Disord 16:329–333. https://doi.org/10.1016/j.parkreldis.2010.02.
007
38. Curtis AR et al (2001) Mutation in the gene encoding ferritin light polypeptide causes domi-
nant adult-onset basal ganglia disease. Nat Genet 28:350–354. https://doi.org/10.1038/ng571
39. Maciel P et al (2005) Neuroferritinopathy: missense mutation in FTL causing early-
onset bilateral pallidal involvement. Neurology 65:603–605. https://doi.org/10.1212/01.wnl.
0000178224.81169.c2
40. Osaki S, Johnson DA, Frieden E (1966) The possible significance of the ferrous oxidase
activity of ceruloplasmin in normal human serum. J Biol Chem 241:2746–2751
41. McNeill A, Pandolfo M, Kuhn J, Shang H, Miyajima H (2008) The neurological presentation
of ceruloplasmin gene mutations. Eur Neurol 60:200–205. https://doi.org/10.1159/000148691
42. Swaiman KF (1991) Hallervorden-Spatz syndrome and brain iron metabolism. Arch Neurol
48:1285–1293
43. Alberca R, Rafel E, Chinchon I, Vadillo J, Navarro A (1987) Late onset parkinsonian syndrome
in Hallervorden-Spatz disease. J Neurol Neurosurg Psychiatry 50:1665–1668
44. Krishnamurthi RV et al (2013) Global and regional burden of first-ever ischaemic and haem-
orrhagic stroke during 1990–2010: findings from the global burden of disease study 2010.
Lancet Global Health 1:E259–E281
45. Langhorne P, Bernhardt J, Kwakkel G (2011) Stroke rehabilitation. Lancet 377:1693–1702.
https://doi.org/10.1016/S0140-6736(11)60325-5
46. Altamura C et al (2009) Ceruloplasmin/transferrin system is related to clinical status in
acute stroke. Stroke: J Cereb Circ 40:1282–1288. https://doi.org/10.1161/STROKEAHA.108.
536714
47. Davalos A et al (1994) Iron-related damage in acute ischemic stroke. Stroke: J Cereb Circ
25:1543–1546
48. Davalos A et al (2000) Body iron stores and early neurologic deterioration in acute cerebral
infarction. Neurology 54:1568–1574
49. Mehdiratta M, Kumar S, Hackney D, Schlaug G, Selim M (2008) Association between serum
ferritin level and perihematoma edema volume in patients with spontaneous intracerebral
hemorrhage. Stroke: J Cereb Circ 39:1165–1170. https://doi.org/10.1161/STROKEAHA.107.
501213
50. Perez de la Ossa, N et al (2010) Iron-related brain damage in patients with intracerebral
hemorrhage. Stroke: J Cereb Circ 41:810–813. https://doi.org/10.1161/strokeaha.109.570168
10 Diagnostics and Treatments of Iron-Related CNS Diseases 191
51. Xiong XY, Wang J, Qian ZM, Yang QW (2014) Iron and intracerebral hemorrhage: from
mechanism to translation. Transl Stroke Res 5:429–441. https://doi.org/10.1007/s12975-013-
0317-7
52. Lou M, Lieb K, Selim M (2009) The relationship between hematoma iron content and per-
ihematoma edema: an MRI study. CerebVascular Dis 27:266–271. https://doi.org/10.1159/
000199464
53. Wu JR, Tuo QZ, Lei P (2018) Ferroptosis, a recent defined form of critical cell death in
neurological disorders. J Mol Neurosci: MN. https://doi.org/10.1007/s12031-018-1155-6
54. Li K, Reichmann H (2016) Role of iron in neurodegenerative diseases. J Neural Transm
123:389–399. https://doi.org/10.1007/s00702-016-1508-7
55. Boddaert N et al (2007) Selective iron chelation in Friedreich ataxia: biologic and clinical
implications. Blood 110:401–408. https://doi.org/10.1182/blood-2006-12-065433
56. Koeppen AH, Morral JA, McComb RD, Feustel PJ (2011) The neuropathology of late-onset
Friedreich’s ataxia. Cerebellum 10:96–103. https://doi.org/10.1007/s12311-010-0235-0
57. Koeppen AH et al (2007) The dentate nucleus in Friedreich’s ataxia: the role of iron-responsive
proteins. Acta Neuropathol 114:163–173. https://doi.org/10.1007/s00401-007-0220-y
58. Rotig A et al (1997) Aconitase and mitochondrial iron-sulphur protein deficiency in Friedreich
ataxia. Nat Genet 17:215–217. https://doi.org/10.1038/ng1097-215
59. Codazzi F et al (2016) Friedreich ataxia-induced pluripotent stem cell-derived neurons show
a cellular phenotype that is corrected by a benzamide HDAC inhibitor. Hum Mol Genet
25:4847–4855. https://doi.org/10.1093/hmg/ddw308
60. Auchere F, Santos R, Planamente S, Lesuisse E, Camadro JM (2008) Glutathione-dependent
redox status of frataxin-deficient cells in a yeast model of Friedreich’s ataxia. Hum Mol Genet
17:2790–2802. https://doi.org/10.1093/hmg/ddn178
61. Wijemanne S, Ondo W (2017) Restless Legs Syndrome: clinical features, diagnosis and a prac-
tical approach to management. Pract Neurol 17:444–452. https://doi.org/10.1136/practneurol-
2017-001762
62. Earley CJ, Barker PB, Horska A, Allen RP (2006) MRI-determined regional brain iron con-
centrations in early- and late-onset restless legs syndrome. Sleep Med 7:458–461. https://doi.
org/10.1016/j.sleep.2005.11.009
63. Rizzo G et al (2013) Low brain iron content in idiopathic restless legs syndrome patients
detected by phase imaging. Mov Disord: Off J Mov Disord Soc 28:1886–1890. https://doi.
org/10.1002/mds.25576
64. Haba-Rubio J et al (2005) Restless legs syndrome and low brain iron levels in patients with
haemochromatosis. J Neurol Neurosurg Psychiatry 76:1009–1010. https://doi.org/10.1136/
jnnp.2003.030536
65. Earley CJ et al (2000) Abnormalities in CSF concentrations of ferritin and transferrin in
restless legs syndrome. Neurology 54:1698–1700
66. Ge Y et al (2007) Quantitative assessment of iron accumulation in the deep gray matter of mul-
tiple sclerosis by magnetic field correlation imaging. AJNR Am J Neuroradiol 28:1639–1644.
https://doi.org/10.3174/ajnr.A0646
67. Haacke EM et al (2009) Characterizing iron deposition in multiple sclerosis lesions using
susceptibility weighted imaging. J Magn Reson Imaging: JMRI 29:537–544. https://doi.org/
10.1002/jmri.21676
68. Hametner S et al (2013) Iron and neurodegeneration in the multiple sclerosis brain. Ann
Neurol 74:848–861. https://doi.org/10.1002/ana.23974
69. Stephenson E, Nathoo N, Mahjoub Y, Dunn JF, Yong VW (2014) Iron in multiple sclerosis:
roles in neurodegeneration and repair. Nat Rev Neurol 10:459–468. https://doi.org/10.1038/
nrneurol.2014.118
70. Weigel KJ, Lynch SG, LeVine SM (2014) Iron chelation and multiple sclerosis. ASN Neuro
6:e00136. https://doi.org/10.1042/AN20130037
71. Benkler C et al (2010) Recent advances in amyotrophic lateral sclerosis research: perspectives
for personalized clinical application. EPMA J 1:343–361. https://doi.org/10.1007/s13167-
010-0026-1
192 H. Xiong et al.
72. Kasarskis EJ, Tandon L, Lovell MA, Ehmann WD (1995) Aluminum, calcium, and iron in
the spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe
mass spectroscopy: a preliminary study. J Neurol Sci 130:203–208
73. Ignjatovic A et al (2012) Inappropriately chelated iron in the cerebrospinal fluid of amy-
otrophic lateral sclerosis patients. Amyotroph Lateral Scler 13:357–362. https://doi.org/10.
3109/17482968.2012.665929
74. Golko-Perez S, Amit T, Bar-Am O, Youdim MB, Weinreb O (2017) A novel iron chelator-
radical scavenger ameliorates motor dysfunction and improves life span and mitochondrial
biogenesis in SOD1(G93A) ALS Mice. Neurotox Res 31:230–244. https://doi.org/10.1007/
s12640-016-9677-6
75. Li X, Jankovic J, Le W (2011) Iron chelation and neuroprotection in neurodegenerative
diseases. J Neural Transm (Vienna) 118:473–477. https://doi.org/10.1007/s00702-010-0518-
0
76. Siciliano G et al (2007) Antioxidant capacity and protein oxidation in cerebrospinal fluid
of amyotrophic lateral sclerosis. J Neurol 254:575–580. https://doi.org/10.1007/s00415-006-
0301-1
77. Goodall EF, Haque MS, Morrison KE (2008) Increased serum ferritin levels in amyotrophic
lateral sclerosis (ALS) patients. J Neurol 255:1652–1656. https://doi.org/10.1007/s00415-
008-0945-0
78. Mizuno Y et al (2006) Transferrin localizes in Bunina bodies in amyotrophic lateral sclerosis.
Acta Neuropathol 112:597–603. https://doi.org/10.1007/s00401-006-0122-4
79. Wang XS et al (2004) Increased incidence of the Hfe mutation in amyotrophic lateral sclerosis
and related cellular consequences. J Neurol Sci 227:27–33. https://doi.org/10.1016/j.jns.2004.
08.003
80. Perls T (2004) Dementia-free centenarians. Exp Gerontol 39:1587–1593. https://doi.org/10.
1016/j.exger.2004.08.015
81. Bush AI (2008) Drug development based on the metals hypothesis of Alzheimer’s disease. J
Alzheimers Dis 15:223–240
82. Ayton S, Lei P, Bush AI (2015) Biometals and their therapeutic implications in Alzheimer’s
disease. Neurotherapeutics 12:109–120. https://doi.org/10.1007/s13311-014-0312-z
83. Crapper McLachlan DR et al (1991) Intramuscular desferrioxamine in patients with
Alzheimer’s disease. Lancet 337:1304–1308
84. Fine JM et al (2012) Intranasal deferoxamine improves performance in radial arm water maze,
stabilizes HIF-1alpha, and phosphorylates GSK3beta in P301L tau transgenic mice. Exp Brain
Res 219:381–390. https://doi.org/10.1007/s00221-012-3101-0
85. Guo C et al (2013) Intranasal deferoxamine reverses iron-induced memory deficits and inhibits
amyloidogenic APP processing in a transgenic mouse model of Alzheimer’s disease. Neuro-
biol Aging 34:562–575. https://doi.org/10.1016/j.neurobiolaging.2012.05.009
86. Guo C et al (2015) Intranasal deferoxamine attenuates synapse loss via up-regulating the
P38/HIF-1alpha pathway on the brain of APP/PS1 transgenic mice. Front Aging Neurosci
7:104. https://doi.org/10.3389/fnagi.2015.00104
87. Di Vaira M et al (2004) Clioquinol, a drug for Alzheimer’s disease specifically interfering with
brain metal metabolism: structural characterization of its zinc(II) and copper(II) complexes.
Inorg Chem 43:3795–3797. https://doi.org/10.1021/ic0494051
88. LeVine H 3rd, Ding Q, Walker JA, Voss RS, Augelli-Szafran CE (2009) Clioquinol and
other hydroxyquinoline derivatives inhibit Abeta(1-42) oligomer assembly. Neurosci Lett
465:99–103. https://doi.org/10.1016/j.neulet.2009.08.002
89. Mancino AM, Hindo SS, Kochi A, Lim MH (2009) Effects of clioquinol on metal-triggered
amyloid-beta aggregation revisited. Inorg Chem 48:9596–9598. https://doi.org/10.1021/
ic9014256
90. Stoppelkamp S et al (2011) In vitro modelling of Alzheimer’s disease: degeneration and cell
death induced by viral delivery of amyloid and tau. Exp Neurol 229:226–237. https://doi.org/
10.1016/j.expneurol.2011.01.018
10 Diagnostics and Treatments of Iron-Related CNS Diseases 193
91. Cherny RA et al (2001) Treatment with a copper-zinc chelator markedly and rapidly inhibits
beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 30:665–676
92. Grossi C et al (2009) Clioquinol decreases amyloid-beta burden and reduces working mem-
ory impairment in a transgenic mouse model of Alzheimer’s disease. J Alzheimers Dis
17:423–440. https://doi.org/10.3233/JAD-2009-1063
93. Ritchie CW et al (2003) Metal-protein attenuation with iodochlorhydroxyquin (clioquinol)
targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical
trial. Archives of neurology 60:1685–1691. https://doi.org/10.1001/archneur.60.12.1685/12/
1685
94. Adlard PA et al (2008) Rapid restoration of cognition in Alzheimer’s transgenic mice with 8-
hydroxy quinoline analogs is associated with decreased interstitial Abeta. Neuron 59:43–55.
https://doi.org/10.1016/j.neuron.2008.06.018
95. Adlard PA et al (2011) Metal ionophore treatment restores dendritic spine density and synaptic
protein levels in a mouse model of Alzheimer’s disease. PLoS ONE 6:e17669. https://doi.org/
10.1371/journal.pone.0017669
96. Crouch PJ et al (2011) The Alzheimer’s therapeutic PBT2 promotes amyloid-beta degradation
and GSK3 phosphorylation via a metal chaperone activity. J Neurochem 119:220–230. https://
doi.org/10.1111/j.1471-4159.2011.07402.x
97. Lannfelt L et al (2008) Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as
a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-
controlled trial. Lancet Neurol 7:779–786. https://doi.org/10.1016/S1474-4422(08)70167-4
98. Faux NG et al (2010) PBT2 rapidly improves cognition in Alzheimer’s Disease: additional
phase II analyses. J Alzheimers Dis 20:509–516. https://doi.org/10.3233/JAD-2010-1390
99. Kwiatkowski A et al (2012) Long-term improvement under deferiprone in a case of neurode-
generation with brain iron accumulation. Parkinsonism Relat Disord 18:110–112. https://doi.
org/10.1016/j.parkreldis.2011.06.024
100. Grolez G et al (2015) Ceruloplasmin activity and iron chelation treatment of patients with
Parkinson’s disease. BMC Neurol 15:74. https://doi.org/10.1186/s12883-015-0331-3
101. Devos D et al (2014) Targeting chelatable iron as a therapeutic modality in Parkinson’s disease.
Antioxid Redox Signal 21:195–210. https://doi.org/10.1089/ars.2013.5593
102. Lei P et al (2015) Clioquinol rescues Parkinsonism and dementia phenotypes of the tau
knockout mouse. Neurobiol Dis 81:168
103. Ayton S et al (2013) Ceruloplasmin dysfunction and therapeutic potential for Parkinson dis-
ease. Ann Neurol 73:554–559. https://doi.org/10.1002/ana.23817
104. Bar-Am O et al (2015) Neuroprotective and neurorestorative activities of a novel iron chelator-
brain selective monoamine oxidase-A/monoamine oxidase-B inhibitor in animal models of
Parkinson’s disease and aging. Neurobiol Aging 36:1529–1542. https://doi.org/10.1016/j.
neurobiolaging.2014.10.026
105. Gal S, Zheng H, Fridkin M, Youdim MB (2005) Novel multifunctional neuroprotective iron
chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selec-
tive brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine
depletion. J Neurochem 95:79–88. https://doi.org/10.1111/j.1471-4159.2005.03341.x
106. Zheng H et al (2005) Novel multifunctional neuroprotective iron chelator-monoamine oxidase
inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant activity, preven-
tion of lipid peroxide formation and monoamine oxidase inhibition. J Neurochem 95:68–78.
https://doi.org/10.1111/j.1471-4159.2005.03340.x
107. Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB (2004) Neuroprotection by
a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats.
Neuropharmacology 46:254–263. https://doi.org/10.1016/S002839080300354X [pii]
108. Kaur D et al (2003) Genetic or pharmacological iron chelation prevents MPTP-induced neu-
rotoxicity in vivo: a novel therapy for Parkinson’s disease. Neuron 37:899–909. https://doi.
org/10.1016/S0896627303001260 [pii]
109. Hasan TF et al (2018) Diagnosis and management of acute ischemic stroke. Mayo Clin Proc
93:523–538. https://doi.org/10.1016/j.mayocp.2018.02.013
194 H. Xiong et al.
110. Iadecola C, Anrather J (2011) Stroke research at a crossroad: asking the brain for directions.
Nat Neurosci 14:1363–1368. https://doi.org/10.1038/nn.2953
111. Selim M (2010) Treatment with the iron chelator, deferoxamine mesylate, alters serum markers
of oxidative stress in stroke patients. Transl Stroke Res 1:35–39. https://doi.org/10.1007/
s12975-009-0001-0
112. Investigators TR (1996) A randomized trial of tirilazad mesylate in patients with acute stroke
(RANTTAS). Stroke: J Cereb Circ 27:1453–1458
113. Haley EC Jr (1998) High-dose tirilazad for acute stroke (RANTTAS II). Stroke: J Cereb Circ
29:1256–1257
114. Hanson LR et al (2009) Intranasal deferoxamine provides increased brain exposure and sig-
nificant protection in rat ischemic stroke. J Pharmacol Exp Ther 330:679–686. https://doi.org/
10.1124/jpet.108.149807
115. Tuo QZ et al (2017) Tau-mediated iron export prevents ferroptotic damage after ischemic
stroke. Mol Psychiatry 22:1520–1530. https://doi.org/10.1038/mp.2017.171
116. Jang YG, Ilodigwe D, Macdonald RL (2009) Metaanalysis of tirilazad mesylate in patients
with aneurysmal subarachnoid hemorrhage. Neurocrit Care 10:141–147. https://doi.org/10.
1007/s12028-008-9147-y
117. Burk K (2017) Friedreich Ataxia: current status and future prospects. Cerebellum & Ataxias
4. https://doi.org/10.1186/s40673-017-0062-x
118. Voncken M, Ioannou P, Delatycki MB (2004) Friedreich ataxia-update on pathogenesis and
possible therapies. Neurogenetics 5:1–8. https://doi.org/10.1007/s10048-003-0170-z
119. Pandolfo M et al (2014) Deferiprone in Friedreich ataxia: a 6-month randomized controlled
trial. Ann Neurol 76:509–521. https://doi.org/10.1002/ana.24248
120. Velasco-Sanchez D et al (2011) Combined therapy with idebenone and deferiprone in patients
with Friedreich’s ataxia. Cerebellum 10:1–8. https://doi.org/10.1007/s12311-010-0212-7
121. Earley CJ, Heckler D, Allen RP (2004) The treatment of restless legs syndrome with intra-
venous iron dextran. Sleep Med 5:231–235. https://doi.org/10.1016/j.sleep.2004.03.002
122. Earley CJ et al (2009) A randomized, double-blind, placebo-controlled trial of intravenous
iron sucrose in restless legs syndrome. Sleep Med 10:206–211. https://doi.org/10.1016/j.sleep.
2007.12.006
123. Pedchenko TV, LeVine SM (1998) Desferrioxamine suppresses experimental allergic
encephalomyelitis induced by MBP in SJL mice. J Neuroimmunol 84:188–197
124. Lynch SG, Peters K, LeVine SM (1996) Desferrioxamine in chronic progressive multiple
sclerosis: a pilot study. Mult Scler 2:157–160. https://doi.org/10.1177/135245859600200306
125. Lynch SG, Fonseca T, LeVine SM (2000) A multiple course trial of desferrioxamine in chronic
progressive multiple sclerosis. Cell Mol Biol (Noisy-le-grand) 46:865–869
126. Creange A, Lefaucheur JP, Balleyguier MO, Galacteros F (2013) Iron depletion induced by
bloodletting and followed by rhEPO administration as a therapeutic strategy in progressive
multiple sclerosis: a pilot, open-label study with neurophysiological measurements. Neuro-
physiol Clin = Clin Neurophysiol 43:303–312. https://doi.org/10.1016/j.neucli.2013.09.004