throbber
Iron: Oxidative Stress and Neurodegeneration
`
`Dev Neurosci 2002;24:188–196
`DOI: 10.1159/000065701
`
`Received: May 2, 2002
`Accepted: June 6, 2002
`
`Brain Iron Metabolism and
`Neurodegenerative Disorders
`
`Jack C. Sipe Pauline Lee Ernest Beutler
`
`Department of Molecular and Experimental Medicine, Scripps Research Institute (MEM-215), La Jolla, Calif., USA
`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`Key Words
`Iron W Metabolism W Gene W Mutation W Single nucleotide
`polymorphism W Neurodegeneration
`
`Abstract
`Iron, an essential element for central nervous system
`(CNS) function, has frequently been found to accumulate
`in brain regions that undergo degeneration in neurologi-
`cal diseases such as Alzheimer disease, Parkinson dis-
`ease, Friedreich ataxia and other disorders. However, the
`precise role of iron in the cause of many neurodegenera-
`tive diseases is unclear. To assist in understanding the
`potential importance of iron in CNS disease, this review
`summarizes the present knowledge in the areas of CNS
`iron metabolism, homeostasis and disregulation of iron
`balance caused by mutations in genes encoding proteins
`involved in iron transport, storage and metabolism. This
`review encompasses neurodegenerative disorders asso-
`ciated with both iron overload and deficiency to highlight
`areas where iron misregulation is likely to be important
`in the pathophysiology of several human brain dis-
`eases.
`
`Copyright © 2002 S. Karger AG, Basel
`
`Introduction
`
`Elemental iron (Fe) is essential for nearly all living
`organisms since it participates in a variety critical meta-
`bolic processes including oxygen transport, electron trans-
`port, DNA synthesis, and redox/non-redox reactions and
`other cell functions [1, 2]. Cellular iron concentrations
`must be tightly controlled due to low iron solubility and
`the ability of Fe to form toxic reactive oxygen interme-
`diates (free radicals) in most biological systems [2].
`In the central nervous system (CNS), iron is necessary
`for many critical metabolic functions. One role of iron in
`the CNS is for the biosynthesis of the neurotransmitters
`dopamine [3] and serotonin [4]. Iron is also important in
`the formation of myelin by oligodendrocytes [5]. In mam-
`mals, brain iron accumulation increases after birth with
`selective uptake in regions undergoing myelination [6]. In
`humans, there is accumulation of iron during adolescence
`in motor areas such as the motor cortex and substantia
`nigra [7, 8]. Iron stores gradually increase until about age
`40 and then remain steady [9]. Many of these iron-depen-
`dent events within the CNS are a function of age, as is the
`case with myelination, or are region specific, as is the case
`with dopamine synthesis in substantia nigra neurons [10].
`In the CNS, iron is also an essential component of
`enzymes of oxidative metabolism, such as those catalyzed
`by the cytochromes and iron-sulfur enzymes. Brain ener-
`
`ABC
`Fax + 41 61 306 12 34
`E-Mail karger@karger.ch
`www.karger.com
`
`© 2002 S. Karger AG, Basel
`
`Accessible online at:
`www.karger.com/journals/dne
`
`Jack C. Sipe, MD
`Department of Molecular and Experimental Medicine
`Scripps Research Institute, 10550 N. Torrey Pines Road
`La Jolla, CA 92037 (USA)
`Tel. +1 818 784 9681, Fax +1 858 784 2083, E-Mail jcsipe@scripps.edu
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 1
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`gy requirements are high, with particular demand for
`ATP as the principal energy source for stable cellular ionic
`gradients, synaptic transmission and distant axoplasmic
`transport [10]. Thus, it is not surprising that Fe-related
`mitochondrial dysfunction has been associated with neu-
`rodegeneration [11]. During CNS oxidative metabolism,
`free radicals are normally produced and metabolized [12],
`but an excess of free Fe results in rapid formation of toxic
`reactive oxygen intermediates that may be one mecha-
`nism of CNS cell death in neurodegeneration [10].
`The purpose of this review is to summarize recent
`advances in the biochemistry, cellular biology and genet-
`ics of iron metabolism as they relate to the critical role of
`iron in normal nervous system function and to proven or
`potential molecular mechanisms of disease in selected
`neurodegenerative disorders. Here, we will focus on the
`molecular control of iron homeostasis in the CNS in both
`health and in neurodegenerative diseases. For more com-
`prehensive reviews of total body iron homeostasis, the
`reader is referred to several recent review articles [1, 2,
`13–15].
`
`Iron Metabolism in the CNS
`
`Iron Uptake and Transport
`Iron uptake in the brain is limited by the blood-brain
`barrier and blood-cerebrospinal fluid (CSF) barrier [16,
`17] maintained by tight junctions between microvascular
`endothelial cells and choroid plexus epithelium. Thus,
`CNS-regulated expression of transport proteins is re-
`quired for uptake of iron [10]. Iron uptake into the brain
`is mediated in part by transferrin receptor expression in
`the endothelial cells and choroid plexus cells [18] or lacto-
`ferrin receptor expression on neurons and microvessels
`[19, 20]. The Fe-transferrin complex or Fe-lactoferrin
`undergoes endocytosis, followed by release of Fe within
`the endothelial cell [10]. Movement of iron from the cyto-
`sol to the extracellular spaces of the CNS involves brain-
`specific proteins. In the CNS, a brain-specific ceruloplas-
`min is expressed as a membrane anchored protein [21]
`and in situ hybridization of CNS tissue reveals abundant
`ceruloplasmin gene expression in specific populations of
`perivascular glial cells associated closely with dopaminer-
`gic melanized neurons in the substantia nigra [22]. Thus,
`ceruloplasmin may play a role in iron export from endo-
`thelial cells into the extracellular spaces or from iron-
`loaded cells such as dopaminergic neurons in the substan-
`tia nigra. The subsequent transport of Fe into other areas
`of the brain is likely mediated by transferrin and non-
`
`transferrin-bound mechanisms [23]. Serum transferrin is
`not found within the CNS unless there is a breakdown of
`the blood-brain barrier [24]. Thus, the CNS must synthe-
`size its own transferrin, a synthesis that is regulated by a
`nervous system-specific promoter [3, 25]. Cellular iron
`uptake may be stimulated by ceruloplasmin ferroxidase
`activity by means of a recently reported trivalent cation-
`specific transport mechanism [26].
`The divalent metal transporter 1 (DMT1, Nramp2)
`has been implicated in transport of iron from endosomes
`and is localized in neurons and oligodendrocytes, which
`show a significantly decreased iron content in the Bel-
`grade rat with a mutation in DMT1/Nramp2 [27]. Alter-
`natively, evidence suggests that Fe may be transported by
`glial cell processes, from astrocyte foot processes that sur-
`round the microvasculature, through the astrocyte cell
`extensions to remote regions [28]. Redistribution studies
`of cerebral iron demonstrate regional movement of iron
`over several weeks in patterns that often suggest the use of
`axonal transport mechanisms to areas of axon projections
`[29]. Ferritin binding, presumably due to receptors, has
`been identified in the human brain [30]. Ferritin-binding
`sites are distributed in iron-rich areas of the brain in con-
`trast to the transferrin receptor distribution which are
`located primarily in the grey matter [30]. As a result, ferri-
`tin receptor binding has been postulated to play a role in
`iron uptake since the localization of ferritin-binding sites
`is better correlated with brain iron accumulation.
`
`Iron Storage
`Ferritin, a cytosolic protein, binds and stores intracel-
`lular iron in most cells of the CNS [12] and prevents it
`from forming reactive oxygen intermediates [1]. In the
`CNS, the synthesis of ferritin takes place in the neuronal
`cell body and holoferritin is transported by axoplasmic
`flow within axons where ferritin may be degraded distally
`within lysosomes [31–33]. Very early in mammalian CNS
`development, iron first appears in microglia [6] and then,
`as brain development continues, ferritin and stored iron
`appear in oligodendroglia in both temporal and regional
`correlation with myelination of axons [5]. The expression
`of ferritin is induced by the presence of excess iron [1].
`Intact human ceruloplasmin appears to be required for
`the incorporation and loading of iron into ferritin [34].
`The concentration of brain ferritin iron may be elevated
`in some neurodegenerative disorders, for example, Alz-
`heimer disease [1] and neuroferrinopathy [35]. Neurome-
`lanin [36] is another major iron storage mechanism in cer-
`tain neuronal systems such as the substantia nigra. Intra-
`neuronal membrane-bound neuromelanin has a strong
`
`Iron and Neurodegeneration
`
`Dev Neurosci 2002;24:188–196
`
`189
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 2
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`chelating ability for iron and may be another mechanism
`of intracellular iron sequestration [37].
`
`Intracellular Iron Utilization
`Iron may be used in the CNS for a wide variety of cellu-
`lar metabolic processes, for example, as a cofactor for the
`enzymes, tyrosine hydroxylase and tryptophan hydroxy-
`lase, in the synthesis of dopamine and serotonin in neu-
`rons or incorporation into iron-sulfur cluster-containing
`proteins [10]. Iron is also essential for the biosynthesis of
`CNS lipids and cholesterol, the important building blocks
`of myelin and as a co-factor for metabolic enzymes in olig-
`odendroglia [5]. The mitochondrion is a particularly criti-
`cal site of intracellular iron utilization since this organelle
`is where iron is incorporated into protoporphyrin IX to
`form heme and where vital non-heme proteins, cyto-
`chromes a, b and c, and cytochrome oxidase required for
`mitochondrial function are also assembled [10]. Mito-
`chondrial iron sequestration has been demonstrated in
`dopamine-challenged astrocytes, providing a possible al-
`ternative pathogenetic mechanism for iron accumulation,
`mitochondrial damage and oxidative injury in neurode-
`generation [38]. Recently, another protein believed to be
`involved in iron metabolism, frataxin, has been localized
`to the mitochondrion in several tissues, including the
`CNS [39–41]. The crystal structure of frataxin has been
`reported [42] and these structural findings predict mecha-
`nisms of protein-protein and protein-iron interactions
`similar to ferritin suggesting that frataxin may serve an
`iron storage function in mitochondria [43].
`
`Iron Recycling and Export
`Most of the iron taken up by the human body is exten-
`sively recycled and reused and the CNS is no exception,
`although little is known about how or if iron exits the
`CNS. Macrophages play a key role in the body by phago-
`cytosis of senescent erythrocytes but red cells are not nor-
`mally present inside the CNS blood-brain barrier except
`in brain injuries [44]. Transferrin-bound Fe in the inter-
`stitial fluid could exit the CNS through the arachnoid villi
`that normally participate in the exit of CSF into the
`venous system [3]. Alternatively, vascular endothelial
`cells could export transferrin-bound iron back into the cir-
`culation [3, 14]. In addition, heme oxygenase (HO) is
`present in the brain in two forms (HO-1 and HO-2) that
`appear to have functions other than heme recycling [45].
`Recent studies of HO distribution demonstrate high lev-
`els of HO-2 in the brain [45] with the highest levels local-
`ized in the substantia nigra [46]. HO-1 appears to be
`involved in mitochondrial iron trapping via the mito-
`
`chondrial transition pore which mediates the transfer of
`non-transferrin iron into mitochondria [38]. HO is upreg-
`ulated in stroke, stress and proinflammatory cytokine
`release which might contribute to brain iron accumula-
`tion in neurodegeneration [47, 48] but one experimental
`brain ischemic injury model suggested that increased HO
`activity does not contribute to iron accumulation [49].
`Consistent with this hypothesis, Ferris et al. [50] showed
`that cells from mice with a targeted deletion of the HO-1
`gene demonstrated reduced iron efflux in HO-1-deficient
`fibroblasts, suggesting a possible mechanism for intracel-
`lular iron accumulation and cell death.
`
`Mechanisms of Iron Homeostasis
`Some fundamentals of CNS iron homeostasis remain
`unknown but clues to these basic mechanisms may be
`uncovered by animal models of iron metabolism dysregu-
`lation produced by targeted genetic deletions. For exam-
`ple, iron regulatory protein-2 (IRP2) gene disruption in
`mice results in iron accumulation in distinctive brain
`regions and is associated with iron overload in degenerat-
`ing neurons and in oligodendrocytes with ubiquitin posi-
`tive inclusions [31]. This leads to a neurodegenerative dis-
`ease in IRP2–/– mice [31] suggesting possible contribu-
`tions to the pathogenesis of comparable human neurologi-
`cal disorders. The function of the hemochromatosis gene,
`HFE, in the brain is unclear but HFE protein has been
`localized to vascular endothelial cells in the cortex and
`cerebellum [51]. However, hereditary hemochromatosis,
`a disorder typically associated with systemic iron over-
`load, has not been associated with excessive brain iron
`levels or neurodegenerative disease. Consistent with this
`notion, Moos et al. [109] examined whether high circula-
`tory levels of iron could cause brain iron accumulation
`and clearly showed that there is exclusion of excess plas-
`ma iron from transfer into the brain.
`
`Neurodegenerative Disorders Associated with
`Increased Total Iron Content
`
`Increased total brain iron content is a feature of some
`neurodegenerative disorders and is thought to be involved
`in the pathophysiology of many of these diseases. In sever-
`al neurodegenerative diseases, an increased concentration
`of brain parenchymal iron has been localized to specific
`brain regions or neuronal systems [3, 43, 52]. Similarly, in
`certain neurodegenerative basal ganglia disorders includ-
`ing Parkinson disease, Huntington disease, Hallervorden-
`Spatz syndrome (neurodegeneration with brain iron accu-
`
`190
`
`Dev Neurosci 2002;24:188–196
`
`Sipe/Lee/Beutler
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 3
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`mulation), Friedreich ataxia, neuroferrinopathy (domi-
`nantly inherited adult-onset basal ganglia disease) and
`hereditary aceruloplasminemia, there is accumulation of
`iron in the basal ganglia [1, 43, 52, 53]. Exactly how
`increased regional or cellular brain iron is involved in dis-
`ease pathophysiology is uncertain but there are several
`possible mechanisms. The potential mechanism most of-
`ten cited is the ability of iron to promote oxidative dam-
`age [3, 43, 54]. It has been proposed that some brain
`regions or specific neuronal groups, i.e., dopaminergic
`neurons, with a high rate of free oxygen radical produc-
`tion would be vulnerable to excessive iron-mediated free
`radicals via the Fenton reaction [55]. Alternatively, the
`presence of high iron levels in Lewy bodies and senile
`plaques in Parkinson disease [56] suggests a possible rela-
`tionship. Iron and oxidative stress might result in the
`damage and aggregation of proteins. In fact, in vitro iron
`has been shown to promote aggregation of ·-synuclein,
`reminiscent of Lewy bodies or amyloid [57]. Hydroxyl
`radicals and other oxygen-free radicals are produced dur-
`ing normal brain metabolism [12, 55] and significantly
`higher plasma levels of hydroxyl radicals have been dem-
`onstrated in Parkinson disease [58], together with in-
`creased lipid peroxidation products in the CSF and brain
`[59, 60], suggesting a role in neurodegeneration. Subse-
`quently, the ubiquitin-proteasome system, essential for
`non-lysosomal catabolism, could fail to adequately clear
`damaged or aggregated proteins in CNS cells [61].
`
`Alzheimer Disease
`Excessive iron accumulation and oxidative damage
`have been associated with Alzheimer disease and may
`contribute to the pathogenesis or disease progression [10].
`In this disease, iron has been associated with senile
`plaques [62], deposition of ß-amyloid peptide fragments
`and abnormal processing of amyloid precursor protein [7,
`63–65]. Analysis of Alzheimer disease brains indicates
`iron accumulation within specific brain regions of se-
`lective vulnerability to neurodegeneration such as the hip-
`pocampus and cerebral cortex [66, 67]. Iron accumulation
`in Alzheimer disease has been suggested as an important
`source of redox-generated free radicals and a contributor
`to oxidative damage in this disease [68]. Neurofibrillary
`tangles and senile plaques contain redox-active metals
`like iron and are major sites for catalytic reactivity [69].
`HO protein immunoreactivity is greatly enhanced in neu-
`rons and astrocytes of the hippocampus and cortex in Alz-
`heimer subjects [70]. Other iron-binding proteins have
`been associated with Alzheimer neurodegeneration in-
`cluding transferrin, ferritin and IRP2. Specifically, brain
`
`tissue from Alzheimer subjects has shown transferrin
`localized to glial cells and melanotransferrin in a subset of
`reactive glia associated with senile plaques [71]. Ferritin-
`containing microglia, ferritin cores and nanoscale iron
`oxides have been demonstrated in senile plaques of Alz-
`heimer brains [63, 72, 73], thus suggesting a possible role
`for ferritin in the disruption of Alzheimer brain iron
`homeostasis. Clearly IRP2 also has an important role in
`neurodegeneration evidenced by IRP2 colocalization in
`senile neurites [74] and by CNS iron overload and degen-
`eration of neurons with associated ubiquitin-positive in-
`clusions in mice with deletion of the IRP2 gene [31].
`Increased expression of melanotransferrin (p97) has
`been evaluated as a biochemical marker for Alzheimer
`disease [75]. Two laboratories have reported that p97 is
`not elevated in other neurodegenerative diseases and may
`be related to overexpression by senile plaque-associated
`reactive microglia [75, 76].
`
`Parkinson Disease
`Impaired iron homeostasis has been demonstrated in
`Parkinson disease and other basal ganglia disorders [77].
`The origin of the increased iron content in Parkinson dis-
`ease is presently unknown. Specifically in Parkinson dis-
`ease, there is increased iron loading of intracellular ferri-
`tin [55] and the iron deposits are found within degenerat-
`ing neurons of the substantia nigra undergoing apoptosis
`and in Lewy bodies [12, 78]. Because Fe is sequestered in
`substantia nigra Lewy bodies [56], increased iron trans-
`port into the substantia nigra and basal ganglia or redistri-
`bution of ferritin or neuromelanin stores is suspected [55].
`It is noteworthy that iron associated with neuromelanin
`significantly accumulates within the substantia nigra in
`Parkinson disease [79] and it has been suggested that this
`mechanism may play a role in the selective neurodegener-
`ation seen in Parkinson disease [12, 80]. HO immunore-
`activity is greatly enhanced in Lewy bodies found in the
`substantia nigra of brains affected with Parkinson disease
`[70]. MPTP, a specific neuronal toxin in the substantia
`nigra that causes parkinsonism, selectively induces HO
`expression in striatal astrocytes [81], thus suggesting a
`possible role of HO in the pathogenesis of Parkinson dis-
`ease. Lactoferrin is structurally similar to transferrin and
`binds iron reversibly but the exact function of lactoferrin
`and lactoferrin receptor in the CNS remain unknown.
`Expression of lactoferrin receptors on neurons and micro-
`vessels has been found to be increased in regions of neu-
`ronal degeneration in the mesencephalon of postmortem
`Parkinson brain tissue and suggests a possible link to iron
`overload in affected brain regions [82].
`
`Iron and Neurodegeneration
`
`Dev Neurosci 2002;24:188–196
`
`191
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 4
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`Table 1. Ceruloplasmin gene
`polymorphisms in Parkinson disease
`
`Location
`
`SNP
`
`Amino
`acid
`
`Control subjects
`n+/n total subjects
`
`Parkinson subjects
`n+/n total subjects
`
`5)-UTR nt-4385)
`UTR nt-16955)
`UTR nt-1936
`
`Ex 9 nt-1652
`
`Ex 11 nt-1950
`
`Ex 17 nt-2991
`
`T → C
`C → T
`A → G
`C → T
`A → C
`T → C
`
`2/2 T/C, C/C
`1/5 C/T
`2/5 A/G
`
`1/5 T/C, 4/5 C/C
`2/8 C/T
`1/8 G/G
`
`T551I
`
`G650G
`
`H997H
`
`0/10
`
`1/10
`
`0/10
`
`3/60
`
`5/60
`
`1/60
`
`Ceruloplasmin gene SNPs and frequencies found in the 5)-untranslated region (5)-UTR)
`and coding exons of white Parkinson disease patients and matched controls. n+ = Number of
`subjects positive for the mutation over the total number (n) of subjects that were genotyped in
`each group. None of the Parkinson subject groups vs. control comparisons were found to be
`statistically significant.
`
`Genes that encode iron-regulatory proteins expressed
`in the CNS have been investigated for a possible role in
`neurodegenerative diseases. Mice with a targeted disrup-
`tion in the IRP2 gene show misregulation of iron metabo-
`lism in the brain and a neurodegenerative disorder with
`significant brain iron overload [31], and the human brain
`distribution of IRP2 colocalizes with redox-active iron
`and the brain lesions of Alzheimer disease [74]. However,
`a new clinical study failed to show an association of any
`natural mutations in the IRP2 gene in patients with spo-
`radic Parkinson disease [83]. Because of a report of
`reduced ferroxidase activity in the CSF of Parkinson dis-
`ease patients [84], we studied the known human cerulo-
`plasmin-coding region single nucleotide polymorphisms
`(SNPs) and promoter region SNPs for association with
`Parkinson disease. With informed consent, we completed
`SNP genotyping on a cohort of up to 60 Parkinson sub-
`jects but the data presented in table 1 do not show any
`significant linkage of ceruloplasmin polymorphisms in
`Parkinson disease compared to matched controls or any
`evidence of SNPs in linkage disequilibrium in Parkinson
`patients [unpublished observations]. Investigations of
`polymorphisms in the 5) flanking region of transferrin
`indicated that the presence of transferrin haplotype 3 in
`white Parkinson patients was in slight excess over the con-
`trol white population suggesting a possible role of trans-
`ferrin SNPs in parkinsonism [85].
`
`Multiple System Atrophy
`Multiple system atrophy (MSA), is an adult-onset neu-
`rodegenerative disorder with parkinsonism, ataxia and
`
`autonomic failure. MSA is characterized by increased
`brain iron [86] and ·-synuclein-containing glial cytoplas-
`mic inclusions [87]. MSA provides a link to a group of
`disorders including Lewy body dementia, Parkinson, and
`Hallervorden-Spatz to which the term ‘synucleinopathy’
`has been applied [87]. A recent brain MRI study in
`patients with MSA demonstrated changes consistent with
`increased ferritin-bound iron and neuromelanin in the
`basal ganglia [88].
`
`Friedreich Ataxia
`Friedreich ataxia, a neurodegenerative disorder, has
`been associated with an intronic trinucleotide repeat ex-
`pansion in the frataxin gene [89] leading to reduced
`mRNA transcription and translation of frataxin protein
`[39]. Patients with Friedreich ataxia demonstrate mito-
`chondrial iron overload [90] and deficiency of iron-sulfur-
`containing enzymes in the CNS [91] In Friedreich ataxia,
`it has been suggested that the decreased levels of frataxin
`and increased iron within mitochondria lead to oxidative
`stress and associated damage of dorsal root ganglion and
`spinal sensory neurons [39, 43].
`
`Aceruloplasminemia
`This autosomal recessive disorder with degeneration of
`the retina and basal ganglia and associated brain iron
`overload results from several loss of function mutations in
`the ceruloplasmin gene [43] and has features of parkin-
`sonism with rigidity and gait disturbance but a lack of
`Parkinson-like tremors. This rare autosomal recessive dis-
`order is estimated to occur at a frequency of 1 per 2 mil-
`
`192
`
`Dev Neurosci 2002;24:188–196
`
`Sipe/Lee/Beutler
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 5
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`lion in Japan [92]. This disorder is associated with several
`mutations in the ceruloplasmin gene. It produces marked
`accumulation of iron in the liver, pancreas, retina, and
`basal ganglia, the latter is thought to contribute to a neu-
`rodegenerative disorder resembling parkinsonism [92].
`CSF from affected patients has shown a threefold elevated
`iron concentration, increased superoxide dismutase activ-
`ity and lipid peroxidation products [59, 93] suggesting a
`direct link between iron overload and oxidative stress as
`one possible causative mechanism.
`
`Hallervorden Spatz
`(neurodegeneration
`syndrome
`Hallervorden-Spatz
`with brain iron accumulation-1, NBIA-1) is a neurodegen-
`erative disorder of childhood associated with iron depos-
`its in the basal ganglia and has been characterized as one
`of the synucleinopathies [94]. The gene responsible for
`NBIA-1 has recently been identified to be the pantothen-
`ate kinase gene (PANK2) that is expressed specifically in
`the brain [95]. Pantothenate kinase is a regulatory enzyme
`that catalyzes the phosphorylation of vitamin B5, panto-
`thenate, but is not directly related to iron metabolism in
`the brain. Thus, an indirect mechanism of secondary
`metabolite accumulation as the cause of basal ganglia iron
`overload in NBIA-1 has been postulated [95].
`
`Neuroferrinopathy
`This dominantly inherited disorder is characterized by
`motor system failure and ferritin-containing inclusions in
`the globus pallidus of the basal ganglia [52]. A mutation
`caused by an insertion of an adenine in the carboxy ter-
`minus of the L chain of ferritin appears to result in a ferri-
`tin molecule with a unique C-terminus. The aberrant fer-
`ritin accumulates in axonal swellings [35] and may be
`involved in the disease pathophysiology. The transport of
`ferritin may be important in the pathophysiology of mice
`lacking IRP2 with neurodegeneration where there is over-
`expression of ferritin H and L chains [52]. Recently, a
`mutation in the iron-responsive element of the Ferritin H
`subunit was associated with tissue iron overload, again
`demonstrating the role of ferritin in inherited iron accu-
`mulation [96].
`
`Other Neurodegenerative Disorders
`Huntington disease and progressive supranuclear palsy
`all show significantly elevated levels of ferritin-associated
`iron in the basal ganglia [72, 73, 78, 79], but the increases
`are present from the onset of the diseases and the relation-
`ship to the origin of these illnesses is uncertain. Multiple
`sclerosis (MS) has been associated with an accumulation
`
`of brain iron [97] although the significance of this finding
`may be only a nonspecific indicator of tissue destruction
`in MS.
`
`Iron Deficiency and CNS Dysfunction
`
`In contrast to CNS iron excess, there have been many
`studies over the years that have examined the issue of iron
`deficiency and CNS dysfunction. The effects of iron defi-
`ciency on the nervous system have been reviewed in the
`past [98, 99] and focused on developmental and intellec-
`tual retardation in experimental animals [100] and on
`developmental and intellectual retardation in humans as
`a consequence of iron deficiency [101, 102]. Neurological
`symptoms in iron-deficient patients have included head-
`aches, papilledema resolving with iron therapy [103], and
`parasthesias. The relationship of intellectual subnormali-
`ty to iron deficiency has been controversial due to the
`complexity of interacting factors [104]. In a study of the
`effects of iron therapy on developmental scores of iron-
`deficient infants, Oski and Honig [105] found improve-
`ment within 1 week of iron supplementation, suggesting
`that elemental Fe deficiency and not the anemia may
`have been responsible for the developmental retardation.
`This could be the result of a lack of functionally critical
`iron-containing proteins such as iron-sulfur proteins, me-
`talloflavoproteins and cytochromes [98]. Studies of iron
`deficiency in animals during development have demon-
`strated low brain iron concentrations and neurobehavio-
`ral abnormalities that persist despite iron treatment after
`weaning [106, 107] suggesting that sufficient brain iron
`content is critical to normal development and behavior
`[107]. Perinatal iron deficiency has also been found to
`decrease cytochrome c oxidase activity in the neonatal rat
`brain hippocampus, demonstrating one mechanism by
`which perinatal iron deficiency reduces neuronal meta-
`bolic activity, especially targeting the brain memory areas
`[108]. Recently, systemic iron deficiency, reduced ferritin
`in the CSF [110] and decreased basal ganglia levels of Fe
`[111] have been documented in some individuals with
`restless legs syndrome suggesting a possible causal link.
`
`Conclusions
`
`Both iron excess and deficiency in the nervous system
`may result in neurodegenerative disorders with motor sys-
`tem and neurobehavioral abnormalities that may have
`devastating or even progressively deteriorating effects
`
`Iron and Neurodegeneration
`
`Dev Neurosci 2002;24:188–196
`
`193
`
`Luitpold Pharmaceuticals, Inc., Ex. 2051, P. 6
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`198.143.32.33 - 3/28/2016 11:36:50 PM
`Downloaded by: L. Kieper - 315380
`
`upon normal brain function. The progressive disability
`seen in many of these disorders is at present largely incur-
`able, although the promise of advances in human genetics
`and molecular biology hold the best chance of identifying
`the cause and either prevention or effective treatment of
`sporadic and inherited neurodegenerative diseases.
`
`Acknowledgments
`
`This is manuscript number 14971-MEM from the Scripps Re-
`search Institute. Supported by the Skaggs Scholars in Clinical Science
`Program from the Scripps Research Institute and grants DK53505–
`02 and RR00833 from the NIH and by the Stein Endowment Fund.
`
`References
`
`1 Lieu P, Heiskala M, Peterson P, Yang Y: The
`roles of iron in health and disease. Mol Aspects
`Med 2001;22:1–87.
`2 Eisenstein R: Iron regulatory proteins and the
`molecular control of mammalian iron metabo-
`lism. Annu Rev Nutr 2000;20:627–662.
`3 Rouault T: Systemic iron metabolism: A re-
`view and implications for brain iron metabo-
`lism. Pediatr Neurol 2001;25:130–137.
`4 D’Sa CM, Arthur RE Jr, States JC, Kuhn DM:
`Tryptophan hydroxylase: Cloning and expres-
`sion of the rat brain enzyme in mammalian
`cells. J Neurochem 1996;67:900–906.
`5 Connor JR, Menzies SL: Relationship of iron
`to oligodendrocytes and myelination. Glia
`1996;17:83–93.
`6 Connor JR, Pavlick G, Karli D, Menzies SL,
`Palmer C: A histochemical study of iron-posi-
`tive cells in the developing rat brain. J Comp
`Neurol 1995;355:111–123.
`7 Connor JR, Snyder BS, Beard JL, Fine RE,
`Mufson EJ: Regional distribution of iron and
`iron-regulatory proteins in the brain in aging
`and Alzheimer’s disease. J Neurosci Res 1992;
`31:327–335.
`8 Aoki S, Okada Y, Nishimura K, Barkovich A,
`Kjos B, Brasch R, Norman D: Normal deposi-
`tion of brain iron in childhood and adoles-
`cence: MR imaging at 1.5 T. Radiology 1989;
`172:381–385.
`9 Bartzokis G, Beckson M, Hance D, Marx P,
`Foster J, Marder S: MR evaluation of age-relat-
`ed increase of brain iron in young adult and
`older normal males. Magn Reson Imaging
`1997;15:29–35.
`10 Connor JR, Menzies SL, Burdo JR, Boyer PJ:
`Iron and iron management proteins in neurobi-
`ology. Pediatr Neurol 2001;25:118–129.
`11 Beal MF: Mitochondrial dysfunction in neu-
`rodegenerative diseases. Biochim Biophys Acta
`1998;1366:211–223.
`12 Faucheux B, Hirsch E: Iron homeostasis and
`Parkinson’s disease (in French). Ann Biol Clin
`(Paris) 1998;56(Spec No):23–30.
`13 Beard JL, Dawson H, Pinero DJ: Iron metabo-
`lism: A comprehensive review. Nutr Rev 1996;
`54:295–317.
`14 Rouault T, Klausner R: Regulation of iron me-
`tabolism in eukaryotes. Curr Top Cell Regul
`1997;35:1–19.
`15 Bothwell T: Overview and mechanisms of iron
`regulation. Nutr Rev 1995;53:237–245.
`
`16 Morris CM, Candy JM, Keith AB, Oakley AE,
`Taylor GA, Pullen RG, Bloxham CA, Gocht A,
`Edwardson JA: Brain iron homeostasis. J Inorg
`Biochem 1992;47:257–265.
`17 Bradbury MW: Transport of iron in the blood-
`brain-cerebrospinal fluid system. J Neurochem
`1997;69:443–454.
`18 Moos T: Immunohistochemical localization of
`intraneuronal transferrin receptor immunore-
`activity in the adult mouse central nervous sys-
`tem. J Comp Neurol 1996;375:675–692.
`19 Kawamata T, Tooyama I, Yamada T, Walker
`DG, McGeer PL: Lactotransferrin immunocy-
`tochemistry in Alzheimer and normal human
`brain. Am J Pathol 1993;142:1574–1585.
`20 Leveugle B, S

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket