Transsulfuration pathway: a targeting neuromodulator in Parkinson’s disease

Abstract The transsulfuration pathway (TSP) is a metabolic pathway involving sulfur transfer from homocysteine to cysteine. Transsulfuration pathway leads to many sulfur metabolites, principally glutathione, H2S, taurine, and cysteine. Key enzymes of the TSP, such as cystathionine β-synthase and cystathionine γ-lyase, are essential regulators at multiple levels in this pathway. TSP metabolites are implicated in many physiological processes in the central nervous system and other tissues. TSP is important in controlling sulfur balance and optimal cellular functions such as glutathione synthesis. Alterations in the TSP and related pathways (transmethylation and remethylation) are altered in several neurodegenerative diseases, including Parkinson’s disease, suggesting their participation in the pathophysiology and progression of these diseases. In Parkinson’s disease many cellular processes are comprised mainly those that regulate redox homeostasis, inflammation, reticulum endoplasmic stress, mitochondrial function, oxidative stress, and sulfur content metabolites of TSP are involved in these damage processes. Current research on the transsulfuration pathway in Parkinson’s disease has primarily focused on the synthesis and function of certain metabolites, particularly glutathione. However, our understanding of the regulation of other metabolites of the transsulfuration pathway, as well as their relationships with other metabolites, and their synthesis regulation in Parkinson´s disease remain limited. Thus, this paper highlights the importance of studying the molecular dynamics in different metabolites and enzymes that affect the transsulfuration in Parkinson’s disease.

[1]  K. Sriram,et al.  Molecular Mechanisms Underlying Neuroinflammation Elicited by Occupational Injuries and Toxicants , 2023, International journal of molecular sciences.

[2]  T. Ezike,et al.  Volatile organic compounds: A proinflammatory activator in autoimmune diseases , 2022, Frontiers in Immunology.

[3]  A. Garcia-Serrano,et al.  Taurine Supplementation as a Neuroprotective Strategy upon Brain Dysfunction in Metabolic Syndrome and Diabetes , 2022, Nutrients.

[4]  T. Bottiglieri,et al.  One-Carbon Metabolism in Alzheimer’s Disease and Parkinson’s Disease Brain Tissue , 2022, Nutrients.

[5]  Nektarios Tavernarakis,et al.  One-Carbon Metabolism: Pulling the Strings behind Aging and Neurodegeneration , 2022, Cells.

[6]  I. Y. Iskusnykh,et al.  Glutathione in Brain Disorders and Aging , 2022, Molecules.

[7]  M. Asanuma,et al.  Glutathione and Related Molecules in Parkinsonism , 2021, International journal of molecular sciences.

[8]  Ray-Jade Chen,et al.  Exercise renovates H2S and Nrf2-related antioxidant pathways to suppress apoptosis in the natural ageing process of male rat cortex , 2021, Biogerontology.

[9]  S. Yao,et al.  Direct Quantification and Visualization of Homocysteine, Cysteine, and Glutathione in Alzheimer's and Parkinson's Disease Model Tissues. , 2021, Analytical chemistry.

[10]  D. Spandidos,et al.  Protective role of taurine against oxidative stress , 2021, Molecular medicine reports.

[11]  A. Tawfik,et al.  Homocysteine and Age-Related Central Nervous System Diseases: Role of Inflammation , 2021, International journal of molecular sciences.

[12]  K. Aoyama Glutathione in the Brain , 2021, International journal of molecular sciences.

[13]  B. Paul Neuroprotective Roles of the Reverse Transsulfuration Pathway in Alzheimer’s Disease , 2021, Frontiers in Aging Neuroscience.

[14]  Ke Wang,et al.  Taurine improves neuron injuries and cognitive impairment in a mouse Parkinson's disease model through inhibition of microglial activation. , 2021, Neurotoxicology.

[15]  Haijun Li,et al.  Role of homocysteine in the development and progression of Parkinson’s disease , 2020, Annals of clinical and translational neurology.

[16]  M. Peana,et al.  The glutathione system in Parkinson’s disease and its progression , 2020, Neuroscience & Biobehavioral Reviews.

[17]  F. Al Mutairi Hyperhomocysteinemia: Clinical Insights , 2020, Journal of central nervous system disease.

[18]  Rana Muhammad Aadil,et al.  Cysteine and homocysteine as biomarker of various diseases , 2020, Food science & nutrition.

[19]  N. Syed,et al.  Taurine Promotes Neurite Outgrowth and Synapse Development of Both Vertebrate and Invertebrate Central Neurons , 2020, Frontiers in Synaptic Neuroscience.

[20]  J. Jankovic,et al.  Parkinson’s disease: etiopathogenesis and treatment , 2020, Journal of Neurology, Neurosurgery, and Psychiatry.

[21]  P. Huang,et al.  Hydrogen Sulfide and Endoplasmic Reticulum Stress: A Potential Therapeutic Target for Central Nervous System Degeneration Diseases , 2020, Frontiers in Pharmacology.

[22]  C. Szabó,et al.  Potential role of the 3-mercaptopyruvate sulfurtransferase (3-MST)-hydrogen sulfide (H2S) pathway in cancer cells. , 2020, Pharmacological research.

[23]  M. Okun,et al.  Diagnosis and Treatment of Parkinson Disease: A Review. , 2020, JAMA.

[24]  Patrik Brundin,et al.  Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. , 2020, Clinics in geriatric medicine.

[25]  Xiangyang Zhu,et al.  Correlations between blood lipid, serum cystatin C, and homocysteine levels in patients with Parkinson's disease , 2019, Psychogeriatrics : the official journal of the Japanese Psychogeriatric Society.

[26]  Ronan M. T. Fleming,et al.  Integrated Analyses of Microbiome and Longitudinal Metabolome Data Reveal Microbial-Host Interactions on Sulfur Metabolism in Parkinson’s Disease , 2019, Cell reports.

[27]  Rubaiya Tabassum,et al.  Potential for therapeutic use of hydrogen sulfide in oxidative stress-induced neurodegenerative diseases , 2019, International journal of medical sciences.

[28]  M. Repici,et al.  DJ-1 in Parkinson’s Disease: Clinical Insights and Therapeutic Perspectives , 2019, Journal of clinical medicine.

[29]  Chaoran Chen,et al.  Roles of taurine in cognitive function of physiology, pathologies and toxication. , 2019, Life sciences.

[30]  F. Mastaglia,et al.  Elevated Serum Homocysteine Levels Have Differential Gender-Specific Associations with Motor and Cognitive States in Parkinson's Disease , 2019, Parkinson's disease.

[31]  D. Choi,et al.  Taurine and its analogs in neurological disorders: Focus on therapeutic potential and molecular mechanisms , 2019, Redox biology.

[32]  S. Snyder,et al.  Regulators of the transsulfuration pathway , 2018, British journal of pharmacology.

[33]  M. De la Fuente,et al.  Lymphoproliferation Impairment and Oxidative Stress in Blood Cells from Early Parkinson’s Disease Patients , 2019, International journal of molecular sciences.

[34]  Guanghui Wang,et al.  Impact of Dopamine Oxidation on Dopaminergic Neurodegeneration. , 2019, ACS chemical neuroscience.

[35]  Hong-Mei Diao,et al.  Association Between MTHFR Genetic Polymorphism and Parkinson’s Disease Susceptibility: A Meta-analysis , 2019, Open medicine.

[36]  R. Sandhir,et al.  Hydrogen sulfide attenuates homocysteine-induced neurotoxicity by preventing mitochondrial dysfunctions and oxidative damage: In vitro and in vivo studies , 2018, Neurochemistry International.

[37]  Siyuan Li,et al.  Protective effects of taurine against inflammation, apoptosis, and oxidative stress in brain injury , 2018, Molecular medicine reports.

[38]  V. Jakovljevic,et al.  Homocysteine and homocysteine-related compounds: an overview of the roles in the pathology of the cardiovascular and nervous systems. , 2018, Canadian journal of physiology and pharmacology.

[39]  S. Chirumbolo,et al.  Metals and Parkinson's Disease: Mechanisms and Biochemical Processes. , 2018, Current medicinal chemistry.

[40]  S. Snyder,et al.  Cysteine Metabolism in Neuronal Redox Homeostasis. , 2018, Trends in pharmacological sciences.

[41]  Yanzhang Li,et al.  Hydrogen sulfide and autophagy: A double edged sword , 2018, Pharmacological research.

[42]  Xiaofang Liu,et al.  Taurine protects dopaminergic neurons in a mouse Parkinson’s disease model through inhibition of microglial M1 polarization , 2018, Cell Death & Disease.

[43]  Liyan Hou,et al.  Taurine protects noradrenergic locus coeruleus neurons in a mouse Parkinson’s disease model by inhibiting microglial M1 polarization , 2018, Amino Acids.

[44]  L. Bobermin,et al.  Homocysteine Induces Glial Reactivity in Adult Rat Astrocyte Cultures , 2017, Molecular Neurobiology.

[45]  J. Bian,et al.  Impaired CBS-H2S signaling axis contributes to MPTP-induced neurodegeneration in a mouse model of Parkinson’s disease , 2018, Brain, Behavior, and Immunity.

[46]  S. Snyder,et al.  Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. , 2017, Biochemical pharmacology.

[47]  T. Montine,et al.  Homocysteine and cognitive function in Parkinson's disease. , 2017, Parkinsonism & related disorders.

[48]  Xiaohu Zhang,et al.  GYY4137, an H2S Slow-Releasing Donor, Prevents Nitrative Stress and α-Synuclein Nitration in an MPTP Mouse Model of Parkinson’s Disease , 2017, Front. Pharmacol..

[49]  Wei-guo Yin,et al.  Neuroprotective effects of lentivirus-mediated cystathionine-beta-synthase overexpression against 6-OHDA-induced parkinson's disease rats , 2017, Neuroscience Letters.

[50]  Chao-shu Tang,et al.  H2S-Induced Sulfhydration: Biological Function and Detection Methodology , 2017, Front. Pharmacol..

[51]  S. Williams,et al.  Glutathione in the human brain: Review of its roles and measurement by magnetic resonance spectroscopy. , 2017, Analytical biochemistry.

[52]  Anthony R. White,et al.  Nexus between mitochondrial function, iron, copper and glutathione in Parkinson's disease , 2017, Neurochemistry International.

[53]  J. Bian,et al.  A New Hope for a Devastating Disease: Hydrogen Sulfide in Parkinson’s Disease , 2017, Molecular Neurobiology.

[54]  T. Yoldas,et al.  Serum glutathione peroxidase, xanthine oxidase, and superoxide dismutase activities and malondialdehyde levels in patients with Parkinson’s disease , 2017, Neurological Sciences.

[55]  O. Tysnes,et al.  Epidemiology of Parkinson’s disease , 2017, Journal of Neural Transmission.

[56]  Jian Li,et al.  Involvement of microRNA-135a-5p in the Protective Effects of Hydrogen Sulfide Against Parkinson's Disease , 2016, Cellular Physiology and Biochemistry.

[57]  Anupom Borah,et al.  L-DOPA-induced hyperhomocysteinemia in Parkinson's disease: Elephant in the room. , 2016, Biochimica et biophysica acta.

[58]  N. Tyagi,et al.  Hydrogen Sulfide Ameliorates Homocysteine-Induced Alzheimer’s Disease-Like Pathology, Blood–Brain Barrier Disruption, and Synaptic Disorder , 2016, Molecular Neurobiology.

[59]  H. Deng,et al.  Association of the MTHFR rs1801131 and rs1801133 variants in sporadic Parkinson’s disease patients , 2016, Neuroscience Letters.

[60]  N. Nagahara,et al.  Identification of H2S3 and H2S produced by 3-mercaptopyruvate sulfurtransferase in the brain , 2015, Scientific Reports.

[61]  Yongsheng Yuan,et al.  Reduced plasma taurine level in Parkinson's disease: association with motor severity and levodopa treatment , 2015, The International journal of neuroscience.

[62]  P. Canto,et al.  Association of the rs1801133 variant in the MTHFR gene and sporadic Parkinson's disease. , 2015, Folia neuropathologica.

[63]  S. Snyder,et al.  Modes of physiologic H2S signaling in the brain and peripheral tissues. , 2015, Antioxidants & redox signaling.

[64]  P. Hrelia,et al.  Neuroprotection by 6-(methylsulfinyl)hexyl isothiocyanate in a 6-hydroxydopamine mouse model of Parkinson׳s disease , 2014, Brain Research.

[65]  Takashi Ito,et al.  Tissue Depletion of Taurine Accelerates Skeletal Muscle Senescence and Leads to Early Death in Mice , 2014, PloS one.

[66]  J. Jia,et al.  Downregulation of cystathionine β-synthase/hydrogen sulfide contributes to rotenone-induced microglia polarization toward M1 type. , 2014, Biochemical and biophysical research communications.

[67]  Phil Hyu Lee,et al.  Endothelial dysfunction and hyperhomocysteinemia in Parkinson's disease: Flow‐mediated dilation study , 2014, Movement disorders : official journal of the Movement Disorder Society.

[68]  B. Tang,et al.  Hypomethylation of SNCA in blood of patients with sporadic Parkinson's disease , 2014, Journal of the Neurological Sciences.

[69]  Seung Yong Park,et al.  Development of a Novel Cysteine Sulfinic Acid Decarboxylase Knockout Mouse: Dietary Taurine Reduces Neonatal Mortality , 2014, Journal of amino acids.

[70]  H. Cho,et al.  Clinical significance of homocysteine (hcy) on dementia in Parkinson's disease (PD). , 2013, Archives of gerontology and geriatrics.

[71]  B. Viskolcz,et al.  Glutathione – Hydroxyl Radical Interaction: A Theoretical Study on Radical Recognition Process , 2013, PloS one.

[72]  M. Smeyne,et al.  Glutathione metabolism and Parkinson's disease. , 2013, Free radical biology & medicine.

[73]  Debabrata Sen,et al.  A ratiometric two-photon fluorescent probe reveals reduction in mitochondrial H2S production in Parkinson's disease gene knockout astrocytes. , 2013, Journal of the American Chemical Society.

[74]  N. Khaper,et al.  Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. , 2013, Antioxidants & redox signaling.

[75]  A. Sparatore,et al.  Therapeutic Effect of Hydrogen Sulfide-Releasing L-Dopa Derivative ACS84 on 6-OHDA-Induced Parkinson’s Disease Rat Model , 2013, PloS one.

[76]  S. Snyder,et al.  Sulfhydration mediates neuroprotective actions of parkin , 2013, Nature Communications.

[77]  T. Guilarte,et al.  Manganese exposure induces α-synuclein aggregation in the frontal cortex of non-human primates. , 2013, Toxicology letters.

[78]  David N Hauser,et al.  Mitochondrial dysfunction and oxidative stress in Parkinson's disease and monogenic parkinsonism , 2013, Neurobiology of Disease.

[79]  A. Schapira,et al.  α-Synuclein and Mitochondrial Dysfunction in Parkinson’s Disease , 2013, Molecular Neurobiology.

[80]  Juan Tang,et al.  The neuroprotection of hydrogen sulfide against MPTP-induced dopaminergic neuron degeneration involves uncoupling protein 2 rather than ATP-sensitive potassium channels. , 2012, Antioxidants & redox signaling.

[81]  M. Gonsebatt,et al.  The role of amino acid transporters in GSH synthesis in the blood–brain barrier and central nervous system , 2012, Neurochemistry International.

[82]  Jang-Yen Wu,et al.  Beneficial effect of taurine on hypoxia- and glutamate-induced endoplasmic reticulum stress pathways in primary neuronal culture , 2012, Amino Acids.

[83]  S. Budrewicz,et al.  The Impact of MRI White Matter Hyperintensities on Dementia in Parkinson's Disease in Relation to the Homocysteine Level and Other Vascular Risk Factors , 2012, Neurodegenerative Diseases.

[84]  J. Bian,et al.  Hydrogen sulfide protects SH-SY5Y cells against 6-hydroxydopamine-induced endoplasmic reticulum stress. , 2012, American journal of physiology. Cell physiology.

[85]  G. Ghanem,et al.  Has removal of excess cysteine led to the evolution of pheomelanin? , 2012, BioEssays : news and reviews in molecular, cellular and developmental biology.

[86]  Wei Zheng,et al.  Regulation of brain iron and copper homeostasis by brain barrier systems: implication in neurodegenerative diseases. , 2012, Pharmacology & therapeutics.

[87]  S. Bearden,et al.  Hyperhomocysteinemia increases permeability of the blood-brain barrier by NMDA receptor-dependent regulation of adherens and tight junctions. , 2011, Blood.

[88]  M. Kaneki,et al.  Inhaled hydrogen sulfide prevents neurodegeneration and movement disorder in a mouse model of Parkinson's disease. , 2011, Antioxidants & redox signaling.

[89]  A. Hayashi‐Takagi,et al.  Glutathione is a physiologic reservoir of neuronal glutamate. , 2011, Biochemical and biophysical research communications.

[90]  Xiao-Qing Tang,et al.  Inhibition of Hydrogen Sulfide Generation Contributes to 1-Methy-4-Phenylpyridinium Ion-Induced Neurotoxicity , 2011, Neurotoxicity Research.

[91]  G. Jameson,et al.  Iron, cysteine and Parkinson’s disease , 2011 .

[92]  T. Chin,et al.  Association of MTHFR, MTR, and MTRR polymorphisms with Parkinson's disease among ethnic Chinese in Taiwan. , 2011, Clinica chimica acta; international journal of clinical chemistry.

[93]  S. Tsuji,et al.  CpG Demethylation Enhances Alpha-Synuclein Expression and Affects the Pathogenesis of Parkinson's Disease , 2010, PloS one.

[94]  J. Bian,et al.  Protective effect of hydrogen sulphide against 6‐OHDA‐induced cell injury in SH‐SY5Y cells involves PKC/PI3K/Akt pathway , 2010, British journal of pharmacology.

[95]  Marc Cruts,et al.  Genetic Etiology of Parkinson Disease Associated with Mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 Genes: A Mutation Update , 2010, Human mutation.

[96]  P. Hluštík,et al.  Effect of entacapone on plasma homocysteine levels in Parkinson’s disease patients , 2010, Neurological Sciences.

[97]  J. Bian,et al.  Neuroprotective effects of hydrogen sulfide on Parkinson’s disease rat models , 2010, Aging cell.

[98]  Chaur-Jong Hu,et al.  Methylenetetrahydrofolate reductase polymorphisms and plasma homocysteine in levodopa-treated and non-treated Parkinson's disease patients , 2009, Journal of the Neurological Sciences.

[99]  U. Dillmann,et al.  Methylation status and neurodegenerative markers in Parkinson disease. , 2009, Clinical chemistry.

[100]  Zhi-Sheng Jiang,et al.  Hydrogen sulfide inhibits MPP(+)-induced apoptosis in PC12 cells. , 2009, Life sciences.

[101]  T. Müller,et al.  Peripheral COMT inhibition prevents levodopa associated homocysteine increase , 2009, Journal of Neural Transmission.

[102]  J. Obeso,et al.  Homocysteine and cognitive impairment in Parkinson's disease: A biochemical, neuroimaging, and genetic study , 2009, Movement disorders : official journal of the Movement Disorder Society.

[103]  Ying Jin,et al.  Protective function of taurine in glutamate‐induced apoptosis in cultured neurons , 2009, Journal of neuroscience research.

[104]  C. Vargas,et al.  Evidence that folic acid deficiency is a major determinant of hyperhomocysteinemia in Parkinson´s disease , 2009, Metabolic Brain Disease.

[105]  Norihiro Shibuya,et al.  3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. , 2009, Antioxidants & redox signaling.

[106]  P. Wong,et al.  Hydrogen Sulfide Inhibits Rotenone-Induced Apoptosis via Preservation of Mitochondrial Function , 2009, Molecular Pharmacology.

[107]  M. Beal,et al.  Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis , 2008, Nature Clinical Practice Neurology.

[108]  J. Andersen,et al.  Oxidative and nitrative protein modifications in Parkinson's disease. , 2008, Free radical biology & medicine.

[109]  G. Nappi,et al.  Homocysteine and Parkinson's disease: A dangerous liaison? , 2007, Journal of the Neurological Sciences.

[110]  V. Ravindranath,et al.  Inhibition of cystathionine-γ-lyase leads to loss of glutathione and aggravation of mitochondrial dysfunction mediated by excitatory amino acid in the CNS , 2007, Neurochemistry International.

[111]  R. Franco,et al.  The central role of glutathione in the pathophysiology of human diseases , 2007, Archives of physiology and biochemistry.

[112]  K. Hasan,et al.  Neuroinflammation, oxidative stress, and the pathogenesis of Parkinson’s disease , 2006, Clinical Neuroscience Research.

[113]  M. Krajinovic,et al.  Homocysteine serum levels and MTHFR C677T genotype in patients with Parkinson's disease, with and without levodopa therapy , 2006, Journal of the Neurological Sciences.

[114]  B. Winblad,et al.  Hyperhomocysteinemia and methylenetetrahydrofolate reductase polymorphism in patients with Parkinson's disease , 2006, Neuroscience Letters.

[115]  D. Selkoe,et al.  Dopamine covalently modifies and functionally inactivates parkin , 2005, Nature Medicine.

[116]  Y. Terayama,et al.  Increase of total homocysteine concentration in cerebrospinal fluid in patients with Alzheimer's disease and Parkinson's disease. , 2005, Life sciences.

[117]  Eun-Sook Y. Lee,et al.  Effects of homocysteine on the dopaminergic system and behavior in rodents. , 2005, Neurotoxicology.

[118]  P. Livrea,et al.  Hyperhomocysteinemia in l‐dopa treated Parkinson's disease patients: effect of cobalamin and folate administration , 2005, European journal of neurology.

[119]  Ying Jin,et al.  Mode of action of taurine as a neuroprotector , 2005, Brain Research.

[120]  P. Livrea,et al.  Plasma homocysteine levels in Parkinson's disease: role of antiparkinsonian medications. , 2005, Parkinsonism & related disorders.

[121]  P. Livrea,et al.  Effects of levodopa and COMT inhibitors on plasma homocysteine in Parkinson's disease patients , 2005, Movement disorders : official journal of the Movement Disorder Society.

[122]  S. Özkan,et al.  Plasma Homocysteine Levels in Pergolide-Treated Parkinson Disease Patients , 2004, Clinical neuropharmacology.

[123]  M. Brosnan,et al.  Methylation demand: a key determinant of homocysteine metabolism. , 2004, Acta biochimica Polonica.

[124]  J. Troncoso,et al.  S-Nitrosylation of Parkin Regulates Ubiquitination and Compromises Parkin's Protective Function , 2004, Science.

[125]  D. Twelves,et al.  Systematic review of incidence studies of Parkinson's disease , 2003, Movement disorders : official journal of the Movement Disorder Society.

[126]  M. Mattson,et al.  Dietary folate deficiency and elevated homocysteine levels endanger dopaminergic neurons in models of Parkinson's disease , 2002, Journal of neurochemistry.

[127]  Barry Halliwell,et al.  Failure of the ubiquitin–proteasome system in Parkinson's disease , 2001, Nature Reviews Neuroscience.

[128]  R. B. Parsons,et al.  Cysteine dioxygenase: Regional localisation of protein and mRNA in rat brain , 2001, Journal of neuroscience research.

[129]  J. B. Justice,et al.  Reaction of oxidized dopamine with endogenous cysteine residues in the human dopamine transporter , 2001, Journal of neurochemistry.

[130]  J. Hirrlinger,et al.  Glutathione metabolism in brain , 2000 .

[131]  K. Tipton,et al.  Taurine‐Induced Attenuation of MPP+ Neurotoxicity In Vitro , 2000, Journal of neurochemistry.

[132]  H. Jakubowski Calcium-dependent Human Serum Homocysteine Thiolactone Hydrolase , 2000, The Journal of Biological Chemistry.

[133]  L. Elferink,et al.  Tyrosine Hydroxylase Is Inactivated by Catechol‐Quinones and Converted to a Redox‐Cycling Quinoprotein , 1999, Journal of neurochemistry.

[134]  K. Jellinger,et al.  The Role of Iron in Neurodegeneration , 1999, Drugs & aging.

[135]  H. Przuntek,et al.  Elevated Plasma Levels of Homocysteine inParkinson’s Disease , 1998, European Neurology.

[136]  A. Lees,et al.  Conjugates of Catecholamines with Cysteine and GSH in Parkinson's Disease: Possible Mechanisms of Formation Involving Reactive Oxygen Species , 1998, Journal of neurochemistry.

[137]  T. Montine,et al.  Neurotoxicity of Endogenous Cysteinylcatechols , 1997, Experimental Neurology.

[138]  G. Dryhurst,et al.  Irreversible Inhibition of Mitochondrial Complex I by 7‐(2‐Aminoethyl)‐3,4‐Dihydro‐5‐Hydroxy‐2H‐1,4‐Benzothiazine‐3‐Carboxylic Acid (DHBT‐1): A Putative Nigral Endotoxin of Relevance to Parkinson's Disease , 1997, Journal of neurochemistry.

[139]  S. Aquilonius,et al.  Levels ofl-MethionineS-Adenosyltranferase Activity in Erythrocytes and Concentrations ofS-Adenosylmethionine andS-Adenosylhomocysteine in Whole Blood of Patients with Parkinson's Disease , 1997, Experimental Neurology.

[140]  B. Shukitt-Hale,et al.  Effect of L-Dopa and the catechol-O-methyltransferase inhibitor Ro 41-0960 on sulfur amino acid metabolites in rats. , 1997, Clinical neuropharmacology.

[141]  L. Ahtee,et al.  Intrastriatal taurine increases striatal extracellular dopamine in a tetrodotoxin-sensitive manner in rats , 1996, Neuroscience Letters.

[142]  G. Dryhurst,et al.  Further insights into the influence of L-cysteine on the oxidation chemistry of dopamine: reaction pathways of potential relevance to Parkinson's disease. , 1996, Chemical research in toxicology.

[143]  G. Omenn,et al.  A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes. , 1995, JAMA.

[144]  M. Marmot,et al.  Survival and cause of death in a cohort of patients with parkinsonism: possible clues to aetiology? , 1995, Journal of neurology, neurosurgery, and psychiatry.

[145]  S. Daniel,et al.  Glutathione‐related enzymes in brain in Parkinson's disease , 1994, Annals of neurology.

[146]  T. Weimbs,et al.  Proteolipid protein (PLP) of CNS myelin: positions of free, disulfide-bonded, and fatty acid thioester-linked cysteine residues and implications for the membrane topology of PLP. , 1992, Biochemistry.

[147]  Peter Riederer,et al.  Transition Metals, Ferritin, Glutathione, and Ascorbic Acid in Parkinsonian Brains , 1989, Journal of neurochemistry.

[148]  M. Palkovits,et al.  Taurine Levels in Discrete Brain Nuclei of Rats , 1986, Journal of neurochemistry.

[149]  K. Kuriyama,et al.  Alteration of amino acids in cerebrospinal fluid from patients with Parkinson's disease and spinocerebellar degeneration , 1986, Acta neurologica Scandinavica.

[150]  A. Meister,et al.  Transport of gamma-glutamyl amino acids: role of glutathione and gamma-glutamyl transpeptidase. , 1979, Proceedings of the National Academy of Sciences of the United States of America.

[151]  D. Straughan,et al.  Synaptic mechanisms in the substantia nigra , 1976, The Journal of pharmacy and pharmacology.

[152]  Dennis W Dickson,et al.  Neuropathology of Parkinson disease. , 2018, Parkinsonism & related disorders.

[153]  J. Volkmann,et al.  Parkinson disease , 2017, Nature Reviews Disease Primers.

[154]  K. Hensley,et al.  Alternative functions of the brain transsulfuration pathway represent an underappreciated aspect of brain redox biochemistry with significant potential for therapeutic engagement. , 2015, Free radical biology & medicine.

[155]  Xiao-Qing Tang,et al.  Neuroprotective effects of hydrogen sulfide and the underlying signaling pathways , 2015, Reviews in the neurosciences.

[156]  A. Antonini,et al.  The MTHFR C677T polymorphism modifies age at onset in Parkinson’s disease , 2013, Neurological Sciences.

[157]  Jang-Yen Wu,et al.  Taurine and central nervous system disorders , 2012, Amino Acids.

[158]  Y. Goto,et al.  Hydrogen sulfide increases glutathione production and suppresses oxidative stress in mitochondria. , 2010, Antioxidants & redox signaling.

[159]  Wen-Jing Li 李文静,et al.  Dose- and time-dependent α-synuclein aggregation induced by ferric iron in SK-N-SH cells , 2010, Neuroscience Bulletin.

[160]  Yu Chen,et al.  The potential protective effects of taurine on coronary heart disease. , 2010, Atherosclerosis.

[161]  Chih-min Yang,et al.  Synergistic effects of homocysteine, S‐adenosylhomocysteine and adenosine on apoptosis in BV‐2 murine microglial cells , 2008, BioFactors.

[162]  P. Blumbergs,et al.  Cellular glutathione peroxidase in human brain: cellular distribution, and its potential role in the degradation of Lewy bodies in Parkinson’s disease and dementia with Lewy bodies , 2008, Acta Neuropathologica.

[163]  M. Brosnan,et al.  Methylation demand and homocysteine metabolism. , 2004, Advances in enzyme regulation.

[164]  R. Diaz-Arrastia,et al.  Elevated plasma homocysteine levels in patients treated with levodopa: association with vascular disease. , 2003, Archives of neurology.

[165]  R. Wolfe,et al.  Homocysteine metabolism. , 1999, Annual review of nutrition.

[166]  W. Tatton,et al.  Etiology and pathogenesis of Parkinson's disease. , 1999, Annual review of neuroscience.

[167]  D. Hoffman,et al.  Relationship between tissue levels of S-adenosylmethionine, S-adenylhomocysteine, and transmethylation reactions. , 1979, Canadian journal of biochemistry.