On the Chemical and Biological Characteristics of Multifunctional Compounds for the Treatment of Parkinson’s Disease

Protein aggregation, mitochondrial dysfunction, iron dyshomeostasis, increased oxidative damage and inflammation are pathognomonic features of Parkinson’s disease (PD) and other neurodegenerative disorders characterized by abnormal iron accumulation. Moreover, the existence of positive feed-back loops between these pathological components, which accelerate, and sometimes make irreversible, the neurodegenerative process, is apparent. At present, the available treatments for PD aim to relieve the symptoms, thus improving quality of life, but no treatments to stop the progression of the disease are available. Recently, the use of multifunctional compounds with the capacity to attack several of the key components of neurodegenerative processes has been proposed as a strategy to slow down the progression of neurodegenerative processes. For the treatment of PD specifically, the necessary properties of new-generation drugs should include mitochondrial destination, the center of iron-reactive oxygen species interaction, iron chelation capacity to decrease iron-mediated oxidative damage, the capacity to quench free radicals to decrease the risk of ferroptotic neuronal death, the capacity to disrupt α-synuclein aggregates and the capacity to decrease inflammatory conditions. Desirable additional characteristics are dopaminergic neurons to lessen unwanted secondary effects during long-term treatment, and the inhibition of the MAO-B and COMPT activities to increase intraneuronal dopamine content. On the basis of the published evidence, in this work, we review the molecular basis underlying the pathological events associated with PD and the clinical trials that have used single-target drugs to stop the progress of the disease. We also review the current information on multifunctional compounds that may be used for the treatment of PD and discuss the chemical characteristics that underlie their functionality. As a projection, some of these compounds or modifications could be used to treat diseases that share common pathology features with PD, such as Friedreich’s ataxia, Multiple sclerosis, Huntington disease and Alzheimer’s disease.

[1]  L. Defebvre,et al.  Trial of Deferiprone in Parkinson's Disease. , 2022, The New England journal of medicine.

[2]  C. A. Stamerra,et al.  Mitochondrial Dysfunction and Cardiovascular Disease: Pathophysiology and Emerging Therapies , 2022, Oxidative medicine and cellular longevity.

[3]  Fudi Wang,et al.  Astrocyte-derived hepcidin controls iron traffic at the blood-brain-barrier via regulating ferroportin 1 of microvascular endothelial cells , 2022, Cell Death & Disease.

[4]  R. Gordon,et al.  Inflammasome Activation in Parkinson’s Disease , 2022, Journal of Parkinson's disease.

[5]  A. Hayes,et al.  LKB1: An emerging therapeutic target for cardiovascular diseases. , 2022, Life sciences.

[6]  M. Sahu,et al.  Age-related Mitochondrial Dysfunction in Parkinson's Disease: New Insights Into the Disease Pathology , 2022, Neuroscience.

[7]  J. Wischhusen,et al.  Neuroinflammation in Parkinson’s Disease – Putative Pathomechanisms and Targets for Disease-Modification , 2022, Frontiers in Immunology.

[8]  Marco A. S. Baptista,et al.  Parkinson’s Disease Drug Therapies in the Clinical Trial Pipeline: 2022 Update , 2022, Journal of Parkinson's disease.

[9]  Margarida M Barroso,et al.  DMT1 bridges endosomes and mitochondria to modulate mitochondrial iron translocation , 2022, bioRxiv.

[10]  M. Tansey,et al.  Inflammation and immune dysfunction in Parkinson disease , 2022, Nature Reviews Immunology.

[11]  S. Ho,et al.  LRRK2 mutant knock-in mouse models: therapeutic relevance in Parkinson's disease , 2022, Translational neurodegeneration.

[12]  J. Duce,et al.  Synthesis, physicochemical characterization and neuroprotective evaluation of novel 1-hydroxypyrazin-2(1H)-one iron chelators in an in vitro cell model of Parkinson's disease , 2022, Dalton transactions.

[13]  M. Mandic,et al.  Antioxidant and neuroprotective activities of selected 2-pyridones: in vitro and in silico study , 2022, Journal of Molecular Structure.

[14]  Qing Wang,et al.  The neuroprotective effects and transdifferentiation of astrocytes into dopaminergic neurons of Ginkgolide K on Parkinson’ disease mice , 2022, Journal of Neuroimmunology.

[15]  J. Morgan,et al.  Low-Dose Niacin Supplementation Improves Motor Function in US Veterans with Parkinson’s Disease: A Single-Center, Randomized, Placebo-Controlled Trial , 2021, Biomedicines.

[16]  N. Davoust,et al.  Chronic Exposure to Paraquat Induces Alpha-Synuclein Pathogenic Modifications in Drosophila , 2021, International journal of molecular sciences.

[17]  Xiaohong Wang,et al.  Ferruginol prevents degeneration of dopaminergic neurons by enhancing clearance of α-synuclein in neuronal cells. , 2021, Fitoterapia.

[18]  Weiguang Zhang,et al.  The Nrf2-NLRP3-caspase-1 axis mediates the neuroprotective effects of Celastrol in Parkinson's disease , 2021, Redox biology.

[19]  C. Miguel,et al.  Antioxidant, anti-inflammatory and neuroprotective actions of resveratrol after experimental nervous system insults. Special focus on the molecular mechanisms involved , 2021, Neurochemistry International.

[20]  J. Menéndez,et al.  Neuroprotective Action of Multitarget 7-Aminophenanthridin-6(5H)-one Derivatives against Metal-Induced Cell Death and Oxidative Stress in SN56 Cells , 2021, ACS chemical neuroscience.

[21]  Jin-Soo Park,et al.  Neuroprotective effects of 2-Heptyl-3-hydroxy-4-quinolone in HT22 mouse hippocampal neuronal cells. , 2021, Bioorganic & medicinal chemistry letters.

[22]  Neena Singh,et al.  Release of Iron-Loaded Ferritin in Sodium Iodate-Induced Model of Age Related Macular Degeneration: An In-Vitro and In-Vivo Study , 2021, Antioxidants.

[23]  T. Fieblinger,et al.  Striatal Control of Movement: A Role for New Neuronal (Sub-) Populations? , 2021, Frontiers in Human Neuroscience.

[24]  R. Gan,et al.  Effects and Mechanisms of Resveratrol on Aging and Age-Related Diseases , 2021, Oxidative medicine and cellular longevity.

[25]  C. Sohn,et al.  Imaging the Substantia Nigra in Parkinson Disease and Other Parkinsonian Syndromes. , 2021, Radiology.

[26]  C. Sőti,et al.  The pleiotropic neuroprotective effects of resveratrol in cognitive decline and Alzheimer’s disease pathology: From antioxidant to epigenetic therapy , 2021, Ageing Research Reviews.

[27]  P. Gao,et al.  ENO1 suppresses cancer cell ferroptosis by degrading the mRNA of iron regulatory protein 1 , 2020, Nature Cancer.

[28]  Changyin Yu,et al.  Signaling mechanisms underlying inhibition of neuroinflammation by resveratrol in neurodegenerative diseases. , 2020, The Journal of nutritional biochemistry.

[29]  L. Dardenne,et al.  Cinnamoyl-N-Acylhydrazone-Donepezil Hybrids: Synthesis and Evaluation of Novel Multifunctional Ligands Against Neurodegenerative Diseases , 2020, Neurochemical Research.

[30]  C. Klein,et al.  Mitochondria and Parkinson’s Disease: Clinical, Molecular, and Translational Aspects , 2020, Journal of Parkinson's disease.

[31]  N. Gretskaya,et al.  The Neuroprotective Effect of N-Docosahexaenoyldopamine on Degenerating Dopaminergic Neurons of the Mesencephalon , 2020, Biology Bulletin.

[32]  Yuqiang Wang,et al.  Neuroprotective and neurogenic effects of novel tetramethylpyrazine derivative T-006 in Parkinson’s disease models through activating the MEF2-PGC1α and BDNF/CREB pathways , 2020, Aging.

[33]  R. Albin,et al.  Current and projected future economic burden of Parkinson’s disease in the U.S. , 2020, npj Parkinson's Disease.

[34]  Y. Ke,et al.  Brain Hepcidin Suppresses Major Pathologies in Experimental Parkinsonism , 2020, iScience.

[35]  Ying Guo,et al.  Design, Synthesis and Biological Evaluation of Novel ( E )‐Hydroxystyryl Aralkyl Sulfones as Neuroprotective Agents , 2020 .

[36]  O. Soukup,et al.  A Perspective on Multi-target Drugs for Alzheimer's Disease. , 2020, Trends in pharmacological sciences.

[37]  Ammara Riaz,et al.  Biochanin A: A novel bioactive multifunctional compound from nature. , 2020, The Science of the total environment.

[38]  N. García,et al.  Mechanisms of mitochondrial DNA escape and its relationship with different metabolic diseases. , 2020, Biochimica et biophysica acta. Molecular basis of disease.

[39]  Benjamin J. Frogley,et al.  Metal coordination to bipyridyl carbynes. , 2020, Dalton transactions.

[40]  J. Elson,et al.  Novel 1-hydroxypyridin-2-one metal chelators prevent and rescue ubiquitin proteasomal-related neuronal injury in an in vitro model of Parkinson’s disease , 2020, Archives of Toxicology.

[41]  U. Das,et al.  Molecular Basis of the Beneficial Actions of Resveratrol. , 2020, Archives of medical research.

[42]  R. Lill,et al.  Mechanisms of Mitochondrial Iron-Sulfur Protein Biogenesis. , 2020, Annual review of biochemistry.

[43]  D. Koziorowski,et al.  Serum levels of hepcidin and interleukin 6 in Parkinson's disease. , 2020, Acta neurobiologiae experimentalis.

[44]  V. Shoshan-Barmatz,et al.  VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease , 2019, Science.

[45]  Aloke K. Dutta,et al.  Targeting alpha synuclein and amyloid beta by a multifunctional, brain-penetrant dopamine D2/D3 agonist D-520: Potential therapeutic application in Parkinson’s disease with dementia , 2019, Scientific Reports.

[46]  Yoichi Chiba,et al.  Immunoreactivities for hepcidin, ferroportin, and hephaestin in astrocytes and choroid plexus epithelium of human brains , 2019, Neuropathology : official journal of the Japanese Society of Neuropathology.

[47]  L. Di,et al.  Tetramethylpyrazine Analogue T-006 Exerts Neuroprotective Effects against 6-Hydroxydopamine-Induced Parkinson's Disease In Vitro and In Vivo , 2019, Oxidative medicine and cellular longevity.

[48]  C. Mawrin,et al.  Mitoferrin-1 is required for brain energy metabolism and hippocampus-dependent memory , 2019, Neuroscience Letters.

[49]  K. Klotz,et al.  The current status of pharmacotherapy for the treatment of Parkinson's disease: transition from single-target to multitarget therapy. , 2019, Drug discovery today.

[50]  Linlin Zhao,et al.  Mitochondrial transcription factor A promotes DNA strand cleavage at abasic sites , 2019, Proceedings of the National Academy of Sciences.

[51]  Siyi Pan,et al.  Isoflavone biochanin A, a novel nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element activator, protects against oxidative damage in HepG2 cells. , 2019, BioFactors.

[52]  Yue Wang,et al.  Discovery of coumarin Mannich base derivatives as multifunctional agents against monoamine oxidase B and neuroinflammation for the treatment of Parkinson's disease. , 2019, European journal of medicinal chemistry.

[53]  W. V. van IJcken,et al.  Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes , 2019, Nature Neuroscience.

[54]  W. Mandemakers,et al.  Parkinson’s disease protein DJ-1 regulates ATP synthase protein components to increase neuronal process outgrowth , 2019, Cell Death & Disease.

[55]  Hao Huang,et al.  Insights on the Multifunctional Activities of Magnolol , 2019, BioMed research international.

[56]  Hui Zhang,et al.  Loss of PINK1 causes age-dependent decrease of dopamine release and mitochondrial dysfunction , 2019, Neurobiology of Aging.

[57]  K. Kalia,et al.  Novel Targets for Parkinson's Disease: Addressing Different Therapeutic Paradigms and Conundrums. , 2019, ACS chemical neuroscience.

[58]  M. Reith,et al.  Design, Synthesis, and Pharmacological Characterization of Carbazole Based Dopamine Agonists as Potential Symptomatic and Neuroprotective Therapeutic Agents for Parkinson's Disease. , 2018, ACS chemical neuroscience.

[59]  M. G. Savelieff,et al.  Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. , 2018, Chemical reviews.

[60]  K. Mohanakumar,et al.  Reinforcing mitochondrial functions in aging brain: An insight into Parkinson's disease therapeutics , 2017, Journal of Chemical Neuroanatomy.

[61]  Eyal Oren,et al.  Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016 , 2017, The Lancet. Neurology.

[62]  M. Roth,et al.  Regulators of hepcidin expression. , 2019, Vitamins and hormones.

[63]  C. Sue,et al.  New insights into the complex role of mitochondria in Parkinson’s disease , 2019, Progress in Neurobiology.

[64]  B. Bloem,et al.  The Emerging Evidence of the Parkinson Pandemic , 2018, Journal of Parkinson's disease.

[65]  A. Hoffbrand,et al.  The Role of Deferiprone in Iron Chelation. , 2018, The New England journal of medicine.

[66]  M. Youdim Monoamine oxidase inhibitors, and iron chelators in depressive illness and neurodegenerative diseases , 2018, Journal of Neural Transmission.

[67]  H. Cai,et al.  Parkin and PINK1 mitigate STING-induced inflammation , 2018, Nature.

[68]  A. Hevener,et al.  New mitochondrial DNA synthesis enables NLRP3 inflammasome activation , 2018, Nature.

[69]  Y. Surova,et al.  Cerebrospinal fluid concentrations of inflammatory markers in Parkinson’s disease and atypical parkinsonian disorders , 2018, Alzheimer's & Dementia.

[70]  Ricardo D. Fernández,et al.  Iron Redox Chemistry Promotes Antiparallel Oligomerization of α-Synuclein. , 2018, Journal of the American Chemical Society.

[71]  T. Südhof,et al.  Cell Biology and Pathophysiology of α-Synuclein. , 2018, Cold Spring Harbor perspectives in medicine.

[72]  Zhenming Hu,et al.  Resveratrol ameliorates autophagic flux to promote functional recovery in rats after spinal cord injury , 2018, Oncotarget.

[73]  Aikseng Ooi,et al.  The Roles of NRF2 in Modulating Cellular Iron Homeostasis , 2017, Antioxidants & redox signaling.

[74]  M. Núñez,et al.  Development of an iron-selective antioxidant probe with protective effects on neuronal function , 2017, PloS one.

[75]  L. Bubacco,et al.  Recent findings on the physiological function of DJ-1: Beyond Parkinson's disease , 2017, Neurobiology of Disease.

[76]  E. H. Howlett,et al.  LRRK2 G2019S-induced mitochondrial DNA damage is LRRK2 kinase dependent and inhibition restores mtDNA integrity in Parkinson’s disease , 2017, Human molecular genetics.

[77]  Souvarish Sarkar,et al.  Mitochondrial impairment in microglia amplifies NLRP3 inflammasome proinflammatory signaling in cell culture and animal models of Parkinson’s disease , 2017, npj Parkinson's Disease.

[78]  J. Bolaños,et al.  Mitochondrial respiratory chain disorganization in Parkinson's disease-relevant PINK1 and DJ1 mutants , 2017, Neurochemistry International.

[79]  Aloke K. Dutta,et al.  D‐512, a novel dopamine D2/3 receptor agonist, demonstrates greater anti‐Parkinsonian efficacy than ropinirole in Parkinsonian rats , 2017, British journal of pharmacology.

[80]  M. Núñez,et al.  Cell death induced by mitochondrial complex I inhibition is mediated by Iron Regulatory Protein 1. , 2017, Biochimica et biophysica acta. Molecular basis of disease.

[81]  J. Andersen,et al.  A novel iron (II) preferring dopamine agonist chelator D-607 significantly suppresses α-syn- and MPTP-induced toxicities in vivo , 2017, Neuropharmacology.

[82]  Khalid Bashir,et al.  Status, supply chain and processing of cocoa - A review , 2017 .

[83]  A. F. Schinder,et al.  αSynuclein control of mitochondrial homeostasis in human-derived neurons is disrupted by mutations associated with Parkinson’s disease , 2017, Scientific Reports.

[84]  Ya-lei Jin,et al.  Chocolate Consumption and Risk of Coronary Heart Disease, Stroke, and Diabetes: A Meta-Analysis of Prospective Studies , 2017, Nutrients.

[85]  U. Förstermann,et al.  Antioxidant effects of resveratrol in the cardiovascular system , 2017, British journal of pharmacology.

[86]  Maneesh C. Patel,et al.  Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease , 2017, Scientific Reports.

[87]  R. Palmiter,et al.  Conditional deletion of Ndufs4 in dopaminergic neurons promotes Parkinson’s disease-like non-motor symptoms without loss of dopamine neurons , 2017, Scientific Reports.

[88]  P. Plastina,et al.  N-Docosahexaenoyl Dopamine, an Endocannabinoid-like Conjugate of Dopamine and the n-3 Fatty Acid Docosahexaenoic Acid, Attenuates Lipopolysaccharide-Induced Activation of Microglia and Macrophages via COX-2. , 2017, ACS chemical neuroscience.

[89]  Olivier Michielin,et al.  SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules , 2017, Scientific Reports.

[90]  Yuqiang Wang,et al.  Neuroprotective Effects and Mechanisms of Action of Multifunctional Agents Targeting Free Radicals, Monoamine Oxidase B and Cholinesterase in Parkinson’s Disease Model , 2017, Journal of Molecular Neuroscience.

[91]  M. Reith,et al.  A Novel Iron(II) Preferring Dopamine Agonist Chelator as Potential Symptomatic and Neuroprotective Therapeutic Agent for Parkinson's Disease. , 2017, ACS chemical neuroscience.

[92]  M. Núñez,et al.  Neuroprotective Effect of a New 7,8-Dihydroxycoumarin-Based Fe2+/Cu2+ Chelator in Cell and Animal Models of Parkinson's Disease. , 2017, ACS chemical neuroscience.

[93]  Raymond Scott Turner,et al.  Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease , 2017, Journal of Neuroinflammation.

[94]  H. Fox,et al.  Loss of Pink1 modulates synaptic mitochondrial bioenergetics in the rat striatum prior to motor symptoms: concomitant complex I respiratory defects and increased complex II‐mediated respiration , 2016, Proteomics. Clinical applications.

[95]  A. Mason,et al.  Erythroid cell mitochondria receive endosomal iron by a "kiss-and-run" mechanism. , 2016, Biochimica et biophysica acta.

[96]  Yong-Xiang Chen,et al.  Phosphorylation induces distinct alpha-synuclein strain formation , 2016, Scientific Reports.

[97]  M. Jimenez-Del-Rio,et al.  Neuroprotective Effect of the LRRK2 Kinase Inhibitor PF-06447475 in Human Nerve-Like Differentiated Cells Exposed to Oxidative Stress Stimuli: Implications for Parkinson’s Disease , 2016, Neurochemical Research.

[98]  V. Dixit,et al.  Inflammasomes: mechanism of assembly, regulation and signalling , 2016, Nature Reviews Immunology.

[99]  Caitlyn W. Barrett,et al.  α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease , 2016, Science Translational Medicine.

[100]  Vincent Zoete,et al.  A BOILED‐Egg To Predict Gastrointestinal Absorption and Brain Penetration of Small Molecules , 2016, ChemMedChem.

[101]  M. Núñez,et al.  Parkinson's Disease: The Mitochondria-Iron Link , 2016, Parkinson's disease.

[102]  Ahmed Negida,et al.  Coenzyme Q10 for Patients with Parkinson's Disease: A Systematic Review and Meta-Analysis. , 2016, CNS & neurological disorders drug targets.

[103]  D. Turnbull,et al.  Mitochondrial DNA Depletion in Respiratory Chain–Deficient Parkinson Disease Neurons , 2016, Annals of neurology.

[104]  Subramanian Rajagopalan,et al.  Regulation of ATP13A2 via PHD2-HIF1α Signaling Is Critical for Cellular Iron Homeostasis: Implications for Parkinson's Disease , 2016, The Journal of Neuroscience.

[105]  J. N. Gattward,et al.  Cocoa and Human Health: From Head to Foot—A Review , 2016, Critical reviews in food science and nutrition.

[106]  Chankyu Park,et al.  Biochanin A Ameliorates Arsenic-Induced Hepato- and Hematotoxicity in Rats , 2016, Molecules.

[107]  M. Reddy,et al.  Hepcidin Plays a Key Role in 6-OHDA Induced Iron Overload and Apoptotic Cell Death in a Cell Culture Model of Parkinson's Disease , 2014, Parkinson's disease.

[108]  Yuan Sheng,et al.  Pre-treatment of rats with ad-hepcidin prevents iron-induced oxidative stress in the brain. , 2016, Free radical biology & medicine.

[109]  Chuen-Mao Yang,et al.  NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review , 2015, Front. Mol. Neurosci..

[110]  O. Isacson,et al.  Fibroblast Biomarkers of Sporadic Parkinson’s Disease and LRRK2 Kinase Inhibition , 2015, Molecular Neurobiology.

[111]  R. Palmiter,et al.  Genetic reduction of mitochondrial complex I function does not lead to loss of dopamine neurons in vivo , 2015, Neurobiology of Aging.

[112]  M. Jensen,et al.  Neurodegeneration with inflammation is accompanied by accumulation of iron and ferritin in microglia and neurons , 2015, Neurobiology of Disease.

[113]  H. P. Adamo,et al.  The Parkinson-associated human P5B-ATPase ATP13A2 protects against the iron-induced cytotoxicity. , 2015, Biochimica et biophysica acta.

[114]  Xingshu Li,et al.  Synthesis and evaluation of selegiline derivatives as monoamine oxidase inhibitor, antioxidant and metal chelator against Alzheimer's disease. , 2015, Bioorganic & medicinal chemistry.

[115]  D. Klenerman,et al.  Aggregated α-synuclein and complex I deficiency: exploration of their relationship in differentiated neurons , 2015, Cell Death and Disease.

[116]  H. Lorenz,et al.  Intramembrane protease PARL defines a negative regulator of PINK1- and PARK2/Parkin-dependent mitophagy , 2015, Autophagy.

[117]  V. Tiranti,et al.  Mitochondria: A crossroads for lipid metabolism defect in neurodegeneration with brain iron accumulation diseases. , 2015, The international journal of biochemistry & cell biology.

[118]  X. Duan,et al.  Neuroprotective effects of ginkgetin against neuroinjury in Parkinson's disease model induced by MPTP via chelating iron , 2015, Free radical research.

[119]  L. Defebvre,et al.  Ceruloplasmin activity and iron chelation treatment of patients with Parkinson’s disease , 2015, BMC Neurology.

[120]  V. Zurawski,et al.  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 , 2015, Neurobiology of Aging.

[121]  P. Chan,et al.  Tea polyphenols alleviate motor impairments, dopaminergic neuronal injury, and cerebral α-synuclein aggregation in MPTP-intoxicated parkinsonian monkeys , 2015, Neuroscience.

[122]  R. Means,et al.  Mitochondrial DNA Stress Primes the Antiviral Innate Immune Response , 2014, Nature.

[123]  J. Obeso,et al.  Clinical and pathological features of Parkinson's disease. , 2015, Current topics in behavioral neurosciences.

[124]  X. Qi,et al.  Threonine 56 phosphorylation of Bcl-2 is required for LRRK2 G2019S-induced mitochondrial depolarization and autophagy. , 2015, Biochimica et biophysica acta.

[125]  Y. Ke,et al.  Hepcidin Suppresses Brain Iron Accumulation by Downregulating Iron Transport Proteins in Iron-Overloaded Rats , 2015, Molecular Neurobiology.

[126]  Z. Xia,et al.  Maneb-induced dopaminergic neuronal death is not affected by loss of mitochondrial complex I activity: results from primary mesencephalic dopaminergic neurons cultured from individual Ndufs4+/+ and Ndufs4−/− mouse embryos , 2014, Neuroreport.

[127]  M. Youdim,et al.  Molecular targets of the multifunctional iron‐chelating drug, M30, in the brains of mouse models of type 2 diabetes mellitus , 2014, British journal of pharmacology.

[128]  K. Brockmann,et al.  Does uncoupling protein 2 expression qualify as marker of disease status in LRRK2-associated Parkinson's disease? , 2014, Antioxidants & redox signaling.

[129]  M. Martinoli,et al.  Resveratrol as a protective molecule for neuroinflammation: a review of mechanisms. , 2014, Current pharmaceutical biotechnology.

[130]  S. Mandel,et al.  The novel multi-target iron chelator, M30 modulates HIF-1α-related glycolytic genes and insulin signaling pathway in the frontal cortex of APP/PS1 Alzheimer's disease mice. , 2014, Current Alzheimer Research.

[131]  A. Mackay-Sim,et al.  Parkinson's disease-associated human ATP13A2 (PARK9) deficiency causes zinc dyshomeostasis and mitochondrial dysfunction , 2014, Human molecular genetics.

[132]  T. Dawson,et al.  Functional interaction of Parkinson's disease-associated LRRK2 with members of the dynamin GTPase superfamily , 2013, Human molecular genetics.

[133]  M. Boska,et al.  Early Expression of Parkinson’s Disease-Related Mitochondrial Abnormalities in PINK1 Knockout Rats , 2014, Molecular Neurobiology.

[134]  L. Defebvre,et al.  Targeting Chelatable Iron as a Therapeutic Modality in Parkinson ’ s Disease , 2016 .

[135]  Victoria Tapia,et al.  Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells , 2013, Journal of neurochemistry.

[136]  Norihiro Suzuki,et al.  Parkin-catalyzed Ubiquitin-Ester Transfer Is Triggered by PINK1-dependent Phosphorylation* , 2013, The Journal of Biological Chemistry.

[137]  Jun Wang,et al.  Pro-inflammatory cytokines modulate iron regulatory protein 1 expression and iron transportation through reactive oxygen/nitrogen species production in ventral mesencephalic neurons. , 2013, Biochimica et biophysica acta.

[138]  J. Mehta,et al.  Oxidant stress in mitochondrial DNA damage, autophagy and inflammation in atherosclerosis , 2013, Scientific Reports.

[139]  M. Kruer The neuropathology of neurodegeneration with brain iron accumulation. , 2013, International review of neurobiology.

[140]  A. Ramírez,et al.  ATP13A2 mutations impair mitochondrial function in fibroblasts from patients with Kufor-Rakeb syndrome , 2012, Neurobiology of Aging.

[141]  E. Giaime,et al.  Loss of DJ-1 Does Not Affect Mitochondrial Respiration but Increases ROS Production and Mitochondrial Permeability Transition Pore Opening , 2012, PloS one.

[142]  Christine Klein,et al.  Pharmacological Rescue of Mitochondrial Deficits in iPSC-Derived Neural Cells from Patients with Familial Parkinson’s Disease , 2012, Science Translational Medicine.

[143]  E. Bézard,et al.  Loss of P-type ATPase ATP13A2/PARK9 function induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration , 2012, Proceedings of the National Academy of Sciences.

[144]  P. Spano,et al.  1B/(−)IRE DMT1 Expression during Brain Ischemia Contributes to Cell Death Mediated by NF-κB/RelA Acetylation at Lys310 , 2012, PloS one.

[145]  M. Youdim,et al.  Neuroprotection by the multitarget iron chelator M30 on age-related alterations in mice , 2012, Mechanisms of Ageing and Development.

[146]  C. Chu,et al.  ATP13A2 regulates mitochondrial bioenergetics through macroautophagy , 2012, Neurobiology of Disease.

[147]  F. Sterky,et al.  Altered dopamine metabolism and increased vulnerability to MPTP in mice with partial deficiency of mitochondrial complex I in dopamine neurons. , 2012, Human molecular genetics.

[148]  M. Núñez,et al.  Iron toxicity in neurodegeneration , 2012, BioMetals.

[149]  Xiongwei Zhu,et al.  LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. , 2012, Human molecular genetics.

[150]  P. Spano,et al.  1 B / ( 2 ) IRE DMT 1 Expression during Brain Ischemia Contributes to Cell Death Mediated by NFk B / RelA Acetylation at Lys 310 , 2012 .

[151]  R. Palmiter,et al.  Mitochondrial complex III stabilizes complex I in the absence of NDUFS4 to provide partial activity. , 2012, Human molecular genetics.

[152]  D. Hinkle,et al.  DJ-1 knock-down impairs astrocyte mitochondrial function , 2011, Neuroscience.

[153]  S. Mandel,et al.  Novel molecular targets of the neuroprotective/neurorescue multimodal iron chelating drug M30 in the mouse brain , 2011, Neuroscience.

[154]  E. Hirsch,et al.  Effect of mitochondrial complex I inhibition on Fe-S cluster protein activity. , 2011, Biochemical and biophysical research communications.

[155]  B. Galy,et al.  Iron Regulatory Protein 1 Outcompetes Iron Regulatory Protein 2 in Regulating Cellular Iron Homeostasis in Response to Nitric Oxide* , 2011, The Journal of Biological Chemistry.

[156]  R. Nussbaum,et al.  Direct Membrane Association Drives Mitochondrial Fission by the Parkinson Disease-associated Protein α-Synuclein*♦ , 2011, The Journal of Biological Chemistry.

[157]  J. Tschopp,et al.  A role for mitochondria in NLRP3 inflammasome activation , 2011, Nature.

[158]  F. B. Davis,et al.  Resveratrol and apoptosis , 2011, Annals of the New York Academy of Sciences.

[159]  Roberta J. Ward,et al.  Brain iron metabolism and its perturbation in neurological diseases , 2011, Journal of Neural Transmission.

[160]  Mark R. Cookson,et al.  The role of leucine-rich repeat kinase 2 (LRRK2) in Parkinson's disease , 2010, Nature Reviews Neuroscience.

[161]  H. Mortiboys,et al.  Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2 , 2010, Neurology.

[162]  Armin Giese,et al.  Inhibition of mitochondrial fusion by α‐synuclein is rescued by PINK1, Parkin and DJ‐1 , 2010, The EMBO journal.

[163]  Wei Li,et al.  Lysosomal proteolysis is the primary degradation pathway for cytosolic ferritin and cytosolic ferritin degradation is necessary for iron exit. , 2010, Antioxidants & redox signaling.

[164]  M. Vila,et al.  Apoptosis‐inducing factor deficiency sensitizes dopaminergic neurons to parkinsonian neurotoxins , 2010, Annals of neurology.

[165]  N. Bonini,et al.  DJ-1 is critical for mitochondrial function and rescues PINK1 loss of function , 2010, Proceedings of the National Academy of Sciences.

[166]  P. Barter,et al.  Evidence That Niacin Inhibits Acute Vascular Inflammation and Improves Endothelial Dysfunction Independent of Changes in Plasma Lipids , 2010, Arteriosclerosis, thrombosis, and vascular biology.

[167]  N. Hattori,et al.  PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy , 2010, The Journal of cell biology.

[168]  M. Reith,et al.  Discovery of 4-(4-(2-((5-Hydroxy-1,2,3,4-tetrahydronaphthalen-2-yl)(propyl)amino)ethyl)piperazin-1-yl)quinolin-8-ol and its analogues as highly potent dopamine D2/D3 agonists and as iron chelator: in vivo activity indicates potential application in symptomatic and neuroprotective therapy for Parkins , 2010, Journal of medicinal chemistry.

[169]  J. Tschopp,et al.  Thioredoxin-interacting protein links oxidative stress to inflammasome activation , 2010, Nature Immunology.

[170]  P. Klivényi,et al.  Novel therapeutic strategies in Parkinson’s disease , 2010, European Journal of Clinical Pharmacology.

[171]  Ted M. Dawson,et al.  PINK1-dependent recruitment of Parkin to mitochondria in mitophagy , 2009, Proceedings of the National Academy of Sciences.

[172]  Hong Jiang,et al.  Up-regulation of divalent metal transporter 1 is involved in 1-methyl-4-phenylpyridinium (MPP+)-induced apoptosis in MES23.5 cells , 2009, Neurobiology of Aging.

[173]  A H V Schapira,et al.  Levodopa in the treatment of Parkinson’s disease , 2009, European journal of neurology.

[174]  R. Lill Function and biogenesis of iron–sulphur proteins , 2009, Nature.

[175]  A. Ariza,et al.  Molecular Pathology of Lewy Body Diseases , 2009, International journal of molecular sciences.

[176]  Xin Zhao,et al.  Mitochondrial localization of DJ‐1 leads to enhanced neuroprotection , 2009, Journal of neuroscience research.

[177]  Z. Qian,et al.  Role of hepcidin in murine brain iron metabolism , 2009, Cellular and Molecular Life Sciences.

[178]  B. Bacon,et al.  Expression of iron-related genes in human brain and brain tumors , 2009, BMC Neuroscience.

[179]  B. Paw,et al.  Regulation of Mitochondrial Iron Import through Differential Turnover of Mitoferrin 1 and Mitoferrin 2 , 2008, Molecular and Cellular Biology.

[180]  M. Beal,et al.  Mitochondrial Approaches for Neuroprotection , 2008, Annals of the New York Academy of Sciences.

[181]  R. Palmiter,et al.  Mitochondrial complex I inhibition is not required for dopaminergic neuron death induced by rotenone, MPP+, or paraquat , 2008, Proceedings of the National Academy of Sciences.

[182]  Jie Shen,et al.  Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress , 2008, Proceedings of the National Academy of Sciences.

[183]  D. Eliezer,et al.  Structural effects of Parkinson's disease linked DJ‐1 mutations , 2008, Protein science : a publication of the Protein Society.

[184]  M. Chesselet,et al.  Animal models of Parkinson’s disease progression , 2008, Acta Neuropathologica.

[185]  Hui Yang,et al.  Environmental toxins and α-synuclein in Parkinson’s disease , 2007, Molecular Neurobiology.

[186]  P. Emson,et al.  Localization of LRRK2 to membranous and vesicular structures in mammalian brain , 2006, Annals of neurology.

[187]  T. Mitsui,et al.  Parkin affects mitochondrial function and apoptosis in neuronal and myogenic cells. , 2006, Biochemical and biophysical research communications.

[188]  O. von Bohlen und Halbach,et al.  Distribution of the iron‐regulating protein hepcidin in the murine central nervous system , 2006, Journal of neuroscience research.

[189]  U. Brunk,et al.  Intralysosomal iron chelation protects against oxidative stress‐induced cellular damage , 2006, The FEBS journal.

[190]  J. Xie,et al.  Parkinson's Disease Brain Mitochondrial Complex I Has Oxidatively Damaged Subunits and Is Functionally Impaired and Misassembled , 2006, The Journal of Neuroscience.

[191]  A. Brownlie,et al.  Mitoferrin is essential for erythroid iron assimilation , 2006, Nature.

[192]  Prasad N. Paradkar,et al.  Nitric oxide transcriptionally down‐regulates specific isoforms of divalent metal transporter (DMT1) via NF‐κB , 2006, Journal of neurochemistry.

[193]  David S. Park,et al.  Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[194]  S. Mandel,et al.  Mechanism of neuroprotective action of the anti-Parkinson drug rasagiline and its derivatives , 2005, Brain Research Reviews.

[195]  M. Kruszewski,et al.  Induction of iron regulatory protein 1 RNA-binding activity by nitric oxide is associated with a concomitant increase in the labile iron pool: implications for DNA damage. , 2005, Biochemical and biophysical research communications.

[196]  M. Youdim,et al.  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 , 2005, Mechanisms of Ageing and Development.

[197]  M. Youdim,et al.  Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer's, Parkinson's, and other neurodegenerative diseases. , 2005, Bioorganic & medicinal chemistry.

[198]  Jerry J Buccafusco,et al.  Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders. , 2005, Trends in pharmacological sciences.

[199]  G. Reynolds,et al.  Increased iron (III) and total iron content in post mortem substantia nigra of parkinsonian brain , 2005, Journal of Neural Transmission.

[200]  Andrew Lees,et al.  Cloning of the Gene Containing Mutations that Cause PARK8-Linked Parkinson's Disease , 2004, Neuron.

[201]  Francesca Zazzeroni,et al.  Ferritin Heavy Chain Upregulation by NF-κB Inhibits TNFα-Induced Apoptosis by Suppressing Reactive Oxygen Species , 2004, Cell.

[202]  Mark A. Wilson,et al.  The Parkinson's disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[203]  D. Koziorowski,et al.  Mössbauer spectroscopy and ELISA studies reveal differences between Parkinson's disease and control substantia nigra. , 2004, Biochimica et biophysica acta.

[204]  Francesca Zazzeroni,et al.  Ferritin heavy chain upregulation by NF-kappaB inhibits TNFalpha-induced apoptosis by suppressing reactive oxygen species. , 2004, Cell.

[205]  Patrizia Rizzu,et al.  Mutations in the DJ-1 Gene Associated with Autosomal Recessive Early-Onset Parkinsonism , 2002, Science.

[206]  G. Minotti,et al.  Nitric oxide and peroxynitrite activate the iron regulatory protein-1 of J774A.1 macrophages by direct disassembly of the Fe-S cluster of cytoplasmic aconitase. , 2002, Biochemistry.

[207]  F. Torti,et al.  Regulation of ferritin genes and protein. , 2002, Blood.

[208]  J. Drapier,et al.  Nitric oxide and peroxynitrite promote complete disruption of the [4Fe-4S] cluster of recombinant human iron regulatory protein 1 , 2002, JBIC Journal of Biological Inorganic Chemistry.

[209]  V. Uversky,et al.  Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. A possible molecular NK between Parkinson's disease and heavy metal exposure. , 2001, The Journal of biological chemistry.

[210]  K. Pantopoulos,et al.  Modulation of Cellular Iron Metabolism by Hydrogen Peroxide , 2001, The Journal of Biological Chemistry.

[211]  N. Yanagisawa [A prospect of treatment for Parkinson's disease in the 21st century]. , 2000, Nihon rinsho. Japanese journal of clinical medicine.

[212]  K. Davies,et al.  Mitochondrial free radical generation, oxidative stress, and aging. , 2000, Free radical biology & medicine.

[213]  H. Ichinose,et al.  Caspase activities and tumor necrosis factor receptor R1 (p55) level are elevated in the substantia nigra from Parkinsonian brain , 2000, Journal of Neural Transmission.

[214]  D. Radisky,et al.  Iron in cytosolic ferritin can be recycled through lysosomal degradation in human fibroblasts. , 1998, The Biochemical journal.

[215]  P. Lansbury,et al.  NACP, a protein implicated in Alzheimer's disease and learning, is natively unfolded. , 1996, Biochemistry.

[216]  J. Beattie,et al.  Novel inhibitors of prolyl 4-hydroxylase. 5. The intriguing structure-activity relationships seen with 2,2'-bipyridine and its 5,5'-dicarboxylic acid derivatives. , 1993, Journal of medicinal chemistry.

[217]  P. Harrison,et al.  Evidence of H- and L-chains have co-operative roles in the iron-uptake mechanism of human ferritin. , 1992, The Biochemical journal.

[218]  C D Marsden,et al.  Alterations in the levels of iron, ferritin and other trace metals in Parkinson's disease and other neurodegenerative diseases affecting the basal ganglia. , 1991, Brain : a journal of neurology.

[219]  J. Lipsick,et al.  Determinants of sequence-specific DNA-binding by p48v-myb. , 1991, Oncogene.

[220]  Y. Agid,et al.  Iron and Aluminum Increase in the Substantia Nigra of Patients with Parkinson's Disease: An X‐Ray Microanalysis , 1991, Journal of neurochemistry.

[221]  J. A. Watkins,et al.  Mobilization of iron from endocytic vesicles. The effects of acidification and reduction. , 1990, The Journal of biological chemistry.

[222]  C. Marsden,et al.  Basal Lipid Peroxidation in Substantia Nigra Is Increased in Parkinson's Disease , 1989, Journal of neurochemistry.

[223]  E. Huehns,et al.  In vivo evaluation of hydroxypyridone iron chelators in a mouse model. , 1987, Acta haematologica.

[224]  S. Markey,et al.  The Neurotoxicity of 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine in the Monkey and Man , 1984, Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques.

[225]  P. Ballard,et al.  Parkinson's disease in a chemist working with 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. , 1983, The New England journal of medicine.