Unraveling the Role of Heme in Neurodegeneration

Heme (iron-protoporphyrin IX) is an essential co-factor involved in several biological processes, including neuronal survival and differentiation. Nevertheless, an excess of free-heme promotes oxidative stress and lipid peroxidation, thus leading to cell death. The toxic properties of heme in the brain have been extensively studied during intracerebral or subarachnoid hemorrhages. Recently, a growing number of neurodegenerative disorders have been associated to alterations of heme metabolism. Hence, the etiology of such diseases remains undefined. The aim of this review is to highlight the neuropathological role of heme and to discuss the major heme-regulated pathways that might be crucial for the survival of neuronal cells. The understanding of the molecular mechanisms linking heme to neurodegeneration will be important for therapeutic purposes.

[1]  V. Ganapathy,et al.  Polarized distribution of heme transporters in retinal pigment epithelium and their regulation in the iron-overload disease hemochromatosis. , 2011, Investigative ophthalmology & visual science.

[2]  M. Wésolowski-Louvel,et al.  Expression of human ANT2 gene in highly proliferative cells: GRBOX, a new transcriptional element, is involved in the regulation of glycolytic ATP import into mitochondria. , 1998, Journal of molecular biology.

[3]  B. Paw,et al.  Haem homeostasis is regulated by the conserved and concerted functions of HRG-1 proteins , 2008, Nature.

[4]  Hiroshi Suzuki,et al.  Heme regulates gene expression by triggering Crm1‐dependent nuclear export of Bach1 , 2004, The EMBO journal.

[5]  B. Stockwell,et al.  Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease , 2017, Cell.

[6]  M. Pérez,et al.  Mitochondrial Dysfunction Contributes to the Pathogenesis of Alzheimer's Disease , 2015, Oxidative medicine and cellular longevity.

[7]  W. Berger,et al.  Next generation sequencing based identification of disease-associated mutations in Swiss patients with retinal dystrophies , 2016, Scientific Reports.

[8]  J. Abkowitz,et al.  A Heme Export Protein Is Required for Red Blood Cell Differentiation and Iron Homeostasis , 2008, Science.

[9]  Oxidative stress and cellular pathologies in Parkinson’s disease , 2017, Molecular Brain.

[10]  Ann Smith,et al.  Mechanisms of neuroprotection by hemopexin: modeling the control of heme and iron homeostasis in brain neurons in inflammatory states , 2013, Journal of neurochemistry.

[11]  L. Carneiro,et al.  Protein aggregation as a cellular response to oxidative stress induced by heme and iron , 2016, Proceedings of the National Academy of Sciences.

[12]  M. Muckenthaler,et al.  Heme controls ferroportin1 (FPN1) transcription involving Bach1, Nrf2 and a MARE/ARE sequence motif at position −7007 of the FPN1 promoter , 2010, Haematologica.

[13]  N. Verpoorten,et al.  Disease mechanisms in hereditary sensory and autonomic neuropathies , 2006, Neurobiology of Disease.

[14]  W. Frey,et al.  A role for heme in Alzheimer's disease: heme binds amyloid beta and has altered metabolism. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[15]  S. Mellgren,et al.  Familial amyotrophic lateral sclerosis , 1980 .

[16]  E. Masliah,et al.  Oxidative stress induces amyloid-like aggregate formation of NACP/α-synuclein in vitro , 1999 .

[17]  K. Choi,et al.  Paraptosis in the anti-cancer arsenal of natural products. , 2016, Pharmacology & therapeutics.

[18]  M. Beal,et al.  Mitochondrial dysfunction in Parkinson's disease , 2016, Journal of neurochemistry.

[19]  S. Tortorelli,et al.  Cutaneous porphyrias part I: epidemiology, pathogenesis, presentation, diagnosis, and histopathology , 2013, International journal of dermatology.

[20]  Gabriela Kalna,et al.  Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase , 2011, Nature.

[21]  P. Hemachandra Reddy,et al.  Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington's disease: implications for selective neuronal damage. , 2011, Human molecular genetics.

[22]  M. Kiernan,et al.  Purple pigments: The pathophysiology of acute porphyric neuropathy , 2011, Clinical Neurophysiology.

[23]  P. Dyck,et al.  Porphyria and its neurologic manifestations. , 2014, Handbook of clinical neurology.

[24]  C. W. Nogueira,et al.  δ-Aminolevulinate Dehydratase Activity is Stimulated in a MPTP Mouse Model of Parkinson’s Disease: Correlation with Myeloperoxidase Activity , 2017, Cellular and Molecular Neurobiology.

[25]  G. Donmez,et al.  Mitochondrial Biology and Neurological Diseases , 2016, Current neuropharmacology.

[26]  L. Silengo,et al.  Hypoxia controls Flvcr1 gene expression in Caco2 cells through HIF2α and ETS1. , 2014, Biochimica et biophysica acta.

[27]  D. Pareyson,et al.  Posterior column ataxia with retinitis pigmentosa coexisting with sensory‐autonomic neuropathy and leukemia due to the homozygous p.Pro221Ser FLVCR1 mutation , 2017, American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics.

[28]  A. Gregory,et al.  Neurodegeneration with brain iron accumulation. , 2005, Folia neuropathologica.

[29]  Peilin Huang,et al.  ABCG2 reduces ROS‐mediated toxicity and inflammation: a potential role in Alzheimer’s disease , 2010, Journal of neurochemistry.

[30]  H. Bonkovsky,et al.  Clinically Important Features of Porphyrin and Heme Metabolism and the Porphyrias , 2014, Metabolites.

[31]  R. Kauppinen,et al.  An update of clinical management of acute intermittent porphyria , 2015 .

[32]  T. Kawarai,et al.  Proteasome impairment in neural cells derived from HMSN-P patient iPSCs , 2017, Molecular Brain.

[33]  A. Eklund,et al.  GATA transcription factors directly regulate the Parkinson's disease-linked gene α-synuclein , 2008, Proceedings of the National Academy of Sciences.

[34]  D. Richardson,et al.  Mitochondrial dysfunction in the neuro-degenerative and cardio-degenerative disease, Friedreich's ataxia , 2017, Neurochemistry International.

[35]  S. Tsuji,et al.  Posterior column ataxia with retinitis pigmentosa in a Japanese family with a novel mutation in FLVCR1 , 2011, neurogenetics.

[36]  T. Stemmler,et al.  Frataxin and Mitochondrial FeS Cluster Biogenesis* , 2010, The Journal of Biological Chemistry.

[37]  L. Silengo,et al.  The heme exporter Flvcr1 regulates expansion and differentiation of committed erythroid progenitors by controlling intracellular heme accumulation , 2015, Haematologica.

[38]  M. Roussel,et al.  Upregulated heme biosynthesis, an exploitable vulnerability in MYCN-driven leukemogenesis. , 2017, JCI Insight.

[39]  J. Cowan,et al.  Frataxin-mediated Iron Delivery to Ferrochelatase in the Final Step of Heme Biosynthesis* , 2004, Journal of Biological Chemistry.

[40]  J. Gal,et al.  Mitochondrial dysfunction in amyotrophic lateral sclerosis. , 2010, Biochimica et biophysica acta.

[41]  H. Handa,et al.  Adenine Nucleotide Translocator Transports Haem Precursors into Mitochondria , 2008, PloS one.

[42]  Dyonne T Hartong,et al.  Retinitis pigmentosa , 2009 .

[43]  K. McNaught,et al.  Proteasomal function is impaired in substantia nigra in Parkinson's disease , 2001, Neuroscience Letters.

[44]  C. Fallet-Bianco,et al.  Mutations in FLVCR2 are associated with proliferative vasculopathy and hydranencephaly-hydrocephaly syndrome (Fowler syndrome). , 2010, American journal of human genetics.

[45]  M. Beal,et al.  Pink1 regulates mitochondrial dynamics through interaction with the fission/fusion machinery , 2008, Proceedings of the National Academy of Sciences.

[46]  P. Tsai,et al.  A novel TFG mutation causes Charcot-Marie-Tooth disease type 2 and impairs TFG function , 2014, Neurology.

[47]  M. Núñez,et al.  Mitochondrial iron homeostasis and its dysfunctions in neurodegenerative disorders. , 2015, Mitochondrion.

[48]  A. Landar,et al.  Hemin causes mitochondrial dysfunction in endothelial cells through promoting lipid peroxidation: the protective role of autophagy. , 2012, American journal of physiology. Heart and circulatory physiology.

[49]  P. Clouston,et al.  A splice-site variant in FLVCR1 produces retinitis pigmentosa without posterior column ataxia , 2018, Ophthalmic genetics.

[50]  P. So,et al.  The Aging of Iron Man , 2018, Front. Aging Neurosci..

[51]  E. Tolosano,et al.  Heme and erythropoieis: more than a structural role , 2014, Haematologica.

[52]  H. D. de Vries,et al.  Nrf2-induced antioxidant protection: a promising target to counteract ROS-mediated damage in neurodegenerative disease? , 2008, Free radical biology & medicine.

[53]  S. Al-Karadaghi,et al.  The Structure of the Complex between Yeast Frataxin and Ferrochelatase , 2016, The Journal of Biological Chemistry.

[54]  R. Morimoto,et al.  Proteostasis and the aging proteome in health and disease. , 2014, The journals of gerontology. Series A, Biological sciences and medical sciences.

[55]  O. Elemento,et al.  Mutations in FLVCR1 cause posterior column ataxia and retinitis pigmentosa. , 2010, American journal of human genetics.

[56]  M. Ristow,et al.  Frataxin deficiency alters heme pathway transcripts and decreases mitochondrial heme metabolites in mammalian cells. , 2005, Human molecular genetics.

[57]  S. Heinemann,et al.  Heme impairs the ball-and-chain inactivation of potassium channels , 2013, Proceedings of the National Academy of Sciences.

[58]  J. Abkowitz,et al.  Identification of a Human Heme Exporter that Is Essential for Erythropoiesis , 2004, Cell.

[59]  D. Richardson,et al.  Elucidation of the mechanism of mitochondrial iron loading in Friedreich's ataxia by analysis of a mouse mutant , 2009, Proceedings of the National Academy of Sciences.

[60]  C. Tailor,et al.  The Fowler Syndrome-Associated Protein FLVCR2 Is an Importer of Heme , 2010, Molecular and Cellular Biology.

[61]  R. Lewis,et al.  Autosomal recessive posterior column ataxia with retinitis pigmentosa caused by novel mutations in the FLVCR1 gene , 2015, The International journal of neuroscience.

[62]  C. Fallet-Bianco,et al.  Fowler syndrome—A clinical, radiological, and pathological study of 14 cases , 2010, American journal of medical genetics. Part A.

[63]  X. Navarro,et al.  Neurotoxicity induced by antineoplastic proteasome inhibitors. , 2014, Neurotoxicology.

[64]  F. Vinchi,et al.  Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes , 2014, Front. Pharmacol..

[65]  R. Gozzelino The Pathophysiology of Heme in the Brain. , 2016, Current Alzheimer research.

[66]  Da-Neng Wang,et al.  Ins and outs of major facilitator superfamily antiporters. , 2008, Annual review of microbiology.

[67]  L. Silengo,et al.  Mutations in the Heme Exporter FLVCR1 Cause Sensory Neurodegeneration with Loss of Pain Perception , 2016, PLoS genetics.

[68]  J. Dichgans,et al.  Oxidative stress in patients with Friedreich ataxia , 2000, Neurology.

[69]  A. Audhya,et al.  Inhibition of TFG function causes hereditary axon degeneration by impairing endoplasmic reticulum structure , 2013, Proceedings of the National Academy of Sciences.

[70]  D. Winge,et al.  Structure, function, and assembly of heme centers in mitochondrial respiratory complexes. , 2012, Biochimica et biophysica acta.

[71]  R. Boldrini,et al.  Proliferative vasculopathy and hydranencephaly–hydrocephaly syndrome or Fowler syndrome: Report of a family and insight into the disease's mechanism , 2018, Molecular genetics & genomic medicine.

[72]  V. Desquiret-Dumas,et al.  Acute intermittent porphyria causes hepatic mitochondrial energetic failure in a mouse model. , 2014, The international journal of biochemistry & cell biology.

[73]  M. Maines,et al.  Overexpression of Heme Oxygenase‐1 Is Neuroprotective in a Model of Permanent Middle Cerebral Artery Occlusion in Transgenic Mice , 1999, Journal of neurochemistry.

[74]  P. Buehler,et al.  Proteasome inhibition and oxidative reactions disrupt cellular homeostasis during heme stress , 2014, Cell Death and Differentiation.

[75]  G. Vanderhoff,et al.  REGULATORY ROLE OF HEME * , 1974, Annals of the New York Academy of Sciences.

[76]  I. Hamza,et al.  Heme Mobilization in Animals: A Metallolipid's Journey. , 2016, Accounts of chemical research.

[77]  T. Stemmler,et al.  Human frataxin: iron and ferrochelatase binding surface. , 2007, Chemical communications.

[78]  F. Han,et al.  The role of endoplasmic reticulum stress in neurodegenerative disease , 2016, Apoptosis.

[79]  C. Vulpe,et al.  Identification of an Intestinal Heme Transporter , 2005, Cell.

[80]  A. Martelli,et al.  Dysregulation of cellular iron metabolism in Friedreich ataxia: from primary iron-sulfur cluster deficit to mitochondrial iron accumulation , 2014, Front. Pharmacol..

[81]  M. Pook,et al.  Animal and cellular models of Friedreich ataxia , 2013, Journal of neurochemistry.

[82]  S. Heinemann,et al.  Metabolic regulation of potassium channels. , 2004, Annual review of physiology.

[83]  P. Lansbury,et al.  A century-old debate on protein aggregation and neurodegeneration enters the clinic , 2006, Nature.

[84]  M. Maes,et al.  The Endoplasmic Reticulum Stress Response in Neuroprogressive Diseases: Emerging Pathophysiological Role and Translational Implications , 2018, Molecular Neurobiology.

[85]  D. Dexter,et al.  Neurodegenerative diseases and therapeutic strategies using iron chelators. , 2015, Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements.

[86]  J. Neurodegenerative Diseases , 1996, GWUMC Department of Biochemistry and Molecular Biology Annual Spring Symposia.

[87]  I. Paulsen,et al.  Major Facilitator Superfamily , 1998, Microbiology and Molecular Biology Reviews.

[88]  D. Manners,et al.  Deficit of in vivo mitochondrial ATP production in patients with Friedreich ataxia. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[89]  A. Nordgren,et al.  Mutations in FLVCR2 associated with Fowler syndrome and survival beyond infancy , 2016, Clinical genetics.

[90]  Emma Lloyd Raven,et al.  The regulatory role of heme in neurons. , 2011, Metallomics : integrated biometal science.

[91]  E. Tolosano,et al.  Mitochondrial Targeting in Neurodegeneration: A Heme Perspective , 2018, Pharmaceuticals.

[92]  M. Beal,et al.  Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases , 2006, Nature.

[93]  P. Patel,et al.  Friedreich's Ataxia: Autosomal Recessive Disease Caused by an Intronic GAA Triplet Repeat Expansion , 1996, Science.

[94]  L. Lue,et al.  ABCG2 Is Upregulated in Alzheimer's Brain with Cerebral Amyloid Angiopathy and May Act as a Gatekeeper at the Blood–Brain Barrier for Aβ1–40 Peptides , 2009, The Journal of Neuroscience.

[95]  C. B. Pickett,et al.  The Nrf2-Antioxidant Response Element Signaling Pathway and Its Activation by Oxidative Stress* , 2009, Journal of Biological Chemistry.

[96]  Peilin Huang,et al.  Abcg2 deficiency augments oxidative stress and cognitive deficits in Tg‐SwDI transgenic mice , 2012, Journal of neurochemistry.

[97]  M. Castro‐Gago,et al.  Congenital Hydranencephalic-Hydrocephalic Syndrome Associated With Mitochondrial Dysfunction , 1999, Journal of child neurology.

[98]  L. Scorrano,et al.  Shaping the role of mitochondria in the pathogenesis of Huntington's disease , 2012, The EMBO journal.

[99]  L. Silengo,et al.  Supplemental Material and Methods , 2014 .

[100]  M. Fleming,et al.  HRG1 is essential for heme transport from the phagolysosome of macrophages during erythrophagocytosis. , 2013, Cell metabolism.

[101]  H. Atamna Heme binding to Amyloid-β peptide: Mechanistic role in Alzheimer's disease , 2006 .

[102]  T. Rouault,et al.  Biogenesis and functions of mammalian iron-sulfur proteins in the regulation of iron homeostasis and pivotal metabolic pathways , 2017, The Journal of Biological Chemistry.

[103]  Youn-Hee Choi,et al.  Repression of adenosine triphosphate–binding cassette transporter ABCG2 by estrogen increases intracellular glutathione in brain endothelial cells following ischemic reperfusion injury , 2018, Neurobiology of Aging.

[104]  K. Bürk Friedreich Ataxia: current status and future prospects , 2017, Cerebellum & Ataxias.

[105]  A. Eschalier,et al.  Potassium channels in neuropathic pain: advances, challenges, and emerging ideas , 2016, Pain.

[106]  Deletion of the hemopexin or heme oxygenase-2 gene aggravates brain injury following stroma-free hemoglobin-induced intracerebral hemorrhage , 2015, Journal of Neuroinflammation.

[107]  F. Supek,et al.  Transcriptional control of AAC3 gene encoding mitochondrial ADP/ATP translocator in Saccharomyces cerevisiae by oxygen, heme and ROX1 factor. , 1993, European journal of biochemistry.

[108]  T. Schwarz Mitochondrial trafficking in neurons. , 2013, Cold Spring Harbor perspectives in biology.

[109]  Xifu Wang,et al.  Induction of heme oxygenase-1 with hemin attenuates hippocampal injury in rats after acute carbon monoxide poisoning. , 2009, Toxicology.

[110]  J. Abkowitz,et al.  Kinetics and Specificity of Feline Leukemia Virus Subgroup C Receptor (FLVCR) Export Function and Its Dependence on Hemopexin* , 2010, The Journal of Biological Chemistry.

[111]  G. Gyapay,et al.  High‐throughput sequencing of a 4.1 Mb linkage interval reveals FLVCR2 deletions and mutations in lethal cerebral vasculopathy , 2010, Human mutation.

[112]  K. Janssens,et al.  Mechanisms of disease in hereditary sensory and autonomic neuropathies , 2012, Nature Reviews Neurology.

[113]  S. Heinemann,et al.  Haem can bind to and inhibit mammalian calcium-dependent Slo1 BK channels , 2003, Nature.

[114]  C. Shaw,et al.  Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal transport to reduce axonal mitochondria content. , 2007, Human molecular genetics.

[115]  M. Hentze,et al.  Iron regulatory protein 1 sustains mitochondrial iron loading and function in frataxin deficiency. , 2015, Cell metabolism.

[116]  R. Lill,et al.  Iron–sulfur cluster biogenesis and trafficking in mitochondria , 2017, The Journal of Biological Chemistry.

[117]  Nada Jabado,et al.  Unexpected allelic heterogeneity and spectrum of mutations in Fowler syndrome revealed by next‐generation exome sequencing , 2010, Human mutation.

[118]  L. Silengo,et al.  Crucial Role of FLVCR1a in the Maintenance of Intestinal Heme Homeostasis. , 2015, Antioxidants & redox signaling.

[119]  M. Zeviani,et al.  Syndromic parkinsonism and dementia associated with OPA 1 missense mutations , 2015, Annals of neurology.

[120]  D. Ito,et al.  TFG-Related Neurologic Disorders: New Insights Into Relationships Between Endoplasmic Reticulum and Neurodegeneration , 2016, Journal of neuropathology and experimental neurology.

[121]  Jose A. Santiago,et al.  Blood Transcriptomic Meta-analysis Identifies Dysregulation of Hemoglobin and Iron Metabolism in Parkinson’ Disease , 2017, Front. Aging Neurosci..

[122]  N. Hirokawa,et al.  The Molecular Motor KIF1A Transports the TrkA Neurotrophin Receptor and Is Essential for Sensory Neuron Survival and Function , 2016, Neuron.

[123]  I. Katona,et al.  Regulation of endoplasmic reticulum turnover by selective autophagy , 2015, Nature.

[124]  M. Castro‐Gago,et al.  Congenital Hydranencephalic-Hydrocephalic Syndrome With Proliferative Vasculopathy: A Possible Relation With Mitochondrial Dysfunction , 2001, Journal of child neurology.

[125]  G. Bennett,et al.  Mitotoxicity in distal symmetrical sensory peripheral neuropathies , 2014, Nature Reviews Neurology.

[126]  S. Schreier,et al.  Hemin-induced lipid membrane disorder and increased permeability: a molecular model for the mechanism of cell lysis. , 1993, Archives of biochemistry and biophysics.

[127]  C. Peers,et al.  Oxidative modulation of K+ channels in the central nervous system in neurodegenerative diseases and aging. , 2015, Antioxidants & redox signaling.

[128]  C. Righy,et al.  Molecular, Cellular and Clinical Aspects of Intracerebral Hemorrhage: Are the Enemies Within? , 2016, Current neuropharmacology.

[129]  D. Butterfield,et al.  Oxidative stress, mitochondrial dysfunction and cellular stress response in Friedreich's ataxia , 2005, Journal of the Neurological Sciences.

[130]  G. Merlo,et al.  The mitochondrial heme exporter FLVCR1b mediates erythroid differentiation. , 2012, The Journal of clinical investigation.

[131]  S. Levi,et al.  Neurodegeneration with brain iron accumulation: update on pathogenic mechanisms , 2014, Front. Pharmacol..

[132]  W. Richard McCombie,et al.  High-Throughput Sequencing , 2011 .

[133]  J. Auwerx,et al.  Pharmacological approaches to restore mitochondrial function , 2013, Nature Reviews Drug Discovery.

[134]  A. Cuervo,et al.  Proteostasis and aging , 2015, Nature Medicine.

[135]  M. Auer-Grumbach Hereditary sensory and autonomic neuropathies. , 2013, Handbook of clinical neurology.

[136]  T. Grune,et al.  Proteasome inhibitors in cancer therapy: Treatment regimen and peripheral neuropathy as a side effect , 2017, Free radical biology & medicine.

[137]  J. Li,et al.  Oxidative Stress and Neurodegenerative Disorders , 2007, International journal of molecular sciences.

[138]  W. Scheper,et al.  The unfolded protein response in neurodegenerative diseases: a neuropathological perspective , 2015, Acta Neuropathologica.

[139]  G. Merlo,et al.  Heme accumulation in endothelial cells impairs angiogenesis by triggering paraptosis , 2017, Cell Death & Differentiation.

[140]  D. Morton,et al.  Posterior column ataxia with retinitis pigmentosa (AXPC1) maps to chromosome 1q31-q32 , 1999, Neurology.

[141]  Sanjay Kumar,et al.  Free heme toxicity and its detoxification systems in human. , 2005, Toxicology letters.

[142]  K. Anderson,et al.  Porphyria Diagnostics—Part 1: A Brief Overview of the Porphyrias , 2015, Current protocols in human genetics.

[143]  D. Marmolino Friedreich's ataxia: Past, present and future , 2011, Brain Research Reviews.

[144]  M. Brand,et al.  Heme regulates the dynamic exchange of Bach1 and NF-E2-related factors in the Maf transcription factor network. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[145]  Thomas L. Williams,et al.  Heme Stabilization of α-Synuclein Oligomers during Amyloid Fibril Formation. , 2015, Biochemistry.

[146]  S. Heinemann,et al.  Oxidative modulation of voltage-gated potassium channels. , 2014, Antioxidants & redox signaling.

[147]  Xiongwei Zhu,et al.  Down-regulation of aminolevulinate synthase, the rate-limiting enzyme for heme biosynthesis in Alzheimer's disease , 2009, Neuroscience Letters.

[148]  J. Schuetz,et al.  Role of ABCG2/BCRP in biology and medicine. , 2006, Annual review of pharmacology and toxicology.

[149]  D. Morton,et al.  An autosomal recessive disorder with posterior column ataxia and retinitis pigmentosa , 1997, Neurology.

[150]  S. Doré,et al.  Heme oxygenase-1 exacerbates early brain injury after intracerebral haemorrhage. , 2007, Brain : a journal of neurology.

[151]  M. Akabas,et al.  Identification of an Intestinal Folate Transporter and the Molecular Basis for Hereditary Folate Malabsorption , 2006, Cell.

[152]  N. Simon,et al.  The neurologic manifestations of the acute porphyrias , 2011, Journal of Clinical Neuroscience.

[153]  Andreas Bracher,et al.  Molecular chaperones in protein folding and proteostasis , 2011, Nature.