[en] Imbalances of iron and dopamine metabolism along with mitochondrial dysfunction have been linked to the pathogenesis of Parkinson's disease (PD). We have previously suggested a direct link between iron homeostasis and dopamine metabolism, as dopamine can increase cellular uptake of iron into macrophages thereby promoting oxidative stress responses. In this study, we investigated the interplay between iron, dopamine, and mitochondrial activity in neuroblastoma SH-SY5Y cells and human induced pluripotent stem cell (hiPSC)-derived dopaminergic neurons differentiated from a healthy control and a PD patient with a mutation in the α-synuclein (SNCA) gene. In SH-SY5Y cells, dopamine treatment resulted in increased expression of the transmembrane iron transporters transferrin receptor 1 (TFR1), ferroportin (FPN), and mitoferrin2 (MFRN2) and intracellular iron accumulation, suggesting that dopamine may promote iron uptake. Furthermore, dopamine supplementation led to reduced mitochondrial fitness including decreased mitochondrial respiration, increased cytochrome c control efficiency, reduced mtDNA copy number and citrate synthase activity, increased oxidative stress and impaired aconitase activity. In dopaminergic neurons derived from a healthy control individual, dopamine showed comparable effects as observed in SH-SY5Y cells. The hiPSC-derived PD neurons harboring an endogenous SNCA mutation demonstrated altered mitochondrial iron homeostasis, reduced mitochondrial capacity along with increased oxidative stress and alterations of tricarboxylic acid cycle linked metabolic pathways compared with control neurons. Importantly, dopamine treatment of PD neurons promoted a rescue effect by increasing mitochondrial respiration, activating antioxidant stress response, and normalizing altered metabolite levels linked to mitochondrial function. These observations provide evidence that dopamine affects iron homeostasis, intracellular stress responses and mitochondrial function in healthy cells, while dopamine supplementation can restore the disturbed regulatory network in PD cells.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
BUOSO, Chiara ; University of Luxembourg ; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy, Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
Seifert, Markus; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria, Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
Lang, Martin; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
GRIFFITH, Corey ; University of Luxembourg > Luxembourg Centre for Systems Biomedicine > Enzymology and Metabolism > Team Carole LINSTER
TALAVERA ANDUJAR, Begona ; University of Luxembourg > Luxembourg Centre for Systems Biomedicine (LCSB) > Environmental Cheminformatics
Rueda, Maria Paulina Castelo; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
Fischer, Christine; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
Doerrier, Carolina; Oroboros Instruments, 6020 Innsbruck, Austria
Talasz, Heribert; Institute of Medical Biochemistry, Protein Core Facility, Biocenter Innsbruck, Medical University of Innsbruck, 6020 Innsbruck, Austria
Zanon, Alessandra; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
Pramstaller, Peter P; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
SCHYMANSKI, Emma ; University of Luxembourg > Luxembourg Centre for Systems Biomedicine (LCSB) > Environmental Cheminformatics
Pichler, Irene; Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy. Electronic address: irene.pichler@eurac.edu
Weiss, Guenter; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria, Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria. Electronic address: Guenter.Weiss@i-med.ac.at
H2020 - 814418 - SinFonia - Synthetic biology-guided engineering of Pseudomonas putida for biofluorination
FnR Project :
FNR11823097 - Microbiomes In One Health, 2017 (01/09/2018-28/02/2025) - Paul Wilmes FNR12341006 - Environmental Cheminformatics To Identify Unknown Chemicals And Their Effects, 2018 (01/10/2018-30/09/2023) - Emma Schymanski
Funders :
FNR - Fonds National de la Recherche EC - European Commission Union Européenne
Aguirre, P., et al. The dopamine metabolite aminochrome inhibits mitochondrial complex I and modifies the expression of iron transporters DMT1 and FPN1. Biometals 25 (2012), 795–803, 10.1007/s10534-012-9525-y.
Ali, M.Y., et al. Mitoferrin, cellular and mitochondrial iron homeostasis. Cells, 11, 2022, 10.3390/cells11213464.
Antonyova, V., et al. Role of mtDNA disturbances in the pathogenesis of Alzheimer's and Parkinson's disease. DNA Repair (Amst), 91-92, 2020, 102871, 10.1016/j.dnarep.2020.102871.
Arreguin, S., et al. Dopamine complexes of iron in the etiology and pathogenesis of Parkinson's disease. J. Inorg. Biochem. 103 (2009), 87–93, 10.1016/j.jinorgbio.2008.09.007.
Beinert, H., Kennedy, M.C., Aconitase, a two-faced protein: enzyme and iron regulatory factor. FASEB J. 7 (1993), 1442–1449, 10.1096/fasebj.7.15.8262329.
Bernal-Conde, L.D., et al. Alpha-synuclein physiology and pathology: a perspective on cellular structures and organelles. Front. Neurosci., 13, 2019, 1399, 10.3389/fnins.2019.01399.
Blauwendraat, C., et al. The genetic architecture of Parkinson's disease. Lancet Neurol. 19 (2020), 170–178, 10.1016/S1474-4422(19)30287-X.
Bolton, E., et al. Pubchemlite for exposomics. Zenodo, 2023, 10.5281/zenodo.7788370.
Burbulla, L.F., et al. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson's disease. Science 357 (2017), 1255–1261, 10.1126/science.aam9080.
Cajka, T., et al. Validating quantitative untargeted lipidomics across nine liquid chromatography-high-resolution mass spectrometry platforms. Anal. Chem. 89 (2017), 12360–12368, 10.1021/acs.analchem.7b03404.
Cardoso, F., Vitamin B12 and Parkinson's disease: what is the relationship?. Mov. Disord. 33 (2018), 702–703, 10.1002/mds.27366.
Castelo Rueda, M.P., et al. Molecular phenotypes of mitochondrial dysfunction in clinically non-manifesting heterozygous PRKN variant carriers. NPJ Parkinsons Dis., 9, 2023, 65, 10.1038/s41531-023-00499-9.
Cerri, S., et al. Endocytic iron trafficking and mitochondria in Parkinson's disease. Int. J. Biochem. Cell Biol. 110 (2019), 70–74, 10.1016/j.biocel.2019.02.009.
Chang, D., et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson's disease risk loci. Nat. Genet. 49 (2017), 1511–1516, 10.1038/ng.3955.
Chartier-Harlin, M.C., et al. Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 364 (2004), 1167–1169, 10.1016/S0140-6736(04)17103-1.
Chen, S.J., et al. Association of fecal and plasma levels of short-chain fatty acids with gut microbiota and clinical severity in patients with Parkinson disease. Neurology 98 (2022), e848–e858, 10.1212/WNL.0000000000013225.
Cheng, R., et al. Mitochondrial iron metabolism and neurodegenerative diseases. Neurotoxicology 88 (2022), 88–101, 10.1016/j.neuro.2021.11.003.
Cheung, Y.-T., et al. Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity research. Neurotoxicology 30 (2009), 127–135, 10.1016/j.neuro.2008.11.001.
Choi, M.L., et al. Pathological structural conversion of alpha-synuclein at the mitochondria induces neuronal toxicity. Nat. Neurosci. 25 (2022), 1134–1148, 10.1038/s41593-022-01140-3.
Ci, Y.Z., et al. Iron overload induced by IRP2 gene knockout aggravates symptoms of Parkinson's disease. Neurochem. Int., 134, 2020, 104657, 10.1016/j.neuint.2019.104657.
Collaborators, G.B.D.P.s.D., Global, regional, and national burden of Parkinson's disease, 1990-2016: a systematic analysis for the global burden of disease study 2016. Lancet Neurol. 17 (2018), 939–953, 10.1016/S1474-4422(18)30295-3.
Collado, M.S., et al. Biochemical and anaplerotic applications of in vitro models of propionic acidemia and methylmalonic acidemia using patient-derived primary hepatocytes. Mol. Genet. Metab. 130 (2020), 183–196, 10.1016/j.ymgme.2020.05.003.
Cronin, S.J.F., et al. The role of iron regulation in immunometabolism and immune-related disease. Front. Mol. Biosci., 6, 2019, 116, 10.3389/fmolb.2019.00116.
Cronin, S.J.F., et al. Crucial neuroprotective roles of the metabolite BH4 in dopaminergic neurons. bioRxiv, 2023, 10.1101/2023.05.08.539795.
David, S., et al. Dysregulation of iron homeostasis in the central nervous system and the role of ferroptosis in neurodegenerative disorders. Antioxid. Redox Signal. 37 (2022), 150–170, 10.1089/ars.2021.0218.
Devos, D., et al. Trial of deferiprone in Parkinson's disease. N. Engl. J. Med. 387 (2022), 2045–2055, 10.1056/NEJMoa2209254.
Di Maio, A., et al. Homeostasis of serine enantiomers is disrupted in the post-mortem caudate putamen and cerebrospinal fluid of living Parkinson's disease patients. Neurobiol. Dis., 184, 2023, 106203, 10.1016/j.nbd.2023.106203.
Dias, V., et al. The role of oxidative stress in Parkinson's disease. J. Parkinsons Dis. 3 (2013), 461–491, 10.3233/JPD-130230.
Dichtl, S., et al. Dopamine promotes cellular iron accumulation and oxidative stress responses in macrophages. Biochem. Pharmacol. 148 (2018), 193–201, 10.1016/j.bcp.2017.12.001.
Dichtl, S., et al. Dopamine is a siderophore-like iron chelator that promotes Salmonella enterica Serovar typhimurium virulence in mice. mBio, 10, 2019, 10.1128/mBio.02624-18.
Doerrier, C., et al. High-resolution FluoRespirometry and OXPHOS protocols for human cells, permeabilized fibers from small biopsies of muscle, and isolated mitochondria. Methods Mol. Biol. 1782 (2018), 31–70, 10.1007/978-1-4939-7831-1_3.
Fischer, C., et al. Dietary Iron overload and Hfe(−/−) related hemochromatosis alter hepatic mitochondrial function. Antioxidants (Basel), 10, 2021, 10.3390/antiox10111818.
Forno, L.S., Neuropathology of Parkinson's disease. J. Neuropathol. Exp. Neurol. 55 (1996), 259–272, 10.1097/00005072-199603000-00001.
Gao, G., et al. Cellular Iron metabolism and regulation. Adv. Exp. Med. Biol. 1173 (2019), 21–32, 10.1007/978-981-13-9589-5_2.
Gilmozzi, V., et al. Generation of hiPSC-derived functional dopaminergic neurons in alginate-based 3D culture. Front. Cell Dev. Biol., 9, 2021, 708389, 10.3389/fcell.2021.708389.
Goetz, D.H., et al. The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron acquisition. Mol. Cell 10 (2002), 1033–1043, 10.1016/s1097-2765(02)00708-6.
Gomez-Giro, G., et al. Synapse alterations precede neuronal damage and storage pathology in a human cerebral organoid model of CLN3-juvenile neuronal ceroid lipofuscinosis. Acta Neuropathol. Commun., 7, 2019, 222, 10.1186/s40478-019-0871-7.
Grunewald, A., et al. New insights into the complex role of mitochondria in Parkinson's disease. Prog. Neurobiol. 177 (2019), 73–93, 10.1016/j.pneurobio.2018.09.003.
Hare, D.J., Double, K.L., Iron and dopamine: a toxic couple. Brain 139 (2016), 1026–1035, 10.1093/brain/aww022.
Haschka, D., et al. Association of mitochondrial iron deficiency and dysfunction with idiopathic restless legs syndrome. Mov. Disord. 34 (2019), 114–123, 10.1002/mds.27482.
He, X., et al. Plasma short-chain fatty acids differences in multiple system atrophy from Parkinson's disease. J. Parkinsons Dis. 11 (2021), 1167–1176, 10.3233/JPD-212604.
Huang, X.T., et al. Iron-induced energy supply deficiency and mitochondrial fragmentation in neurons. J. Neurochem. 147 (2018), 816–830, 10.1111/jnc.14621.
Jager, C., et al. Metabolic profiling and quantification of neurotransmitters in mouse brain by gas chromatography-mass spectrometry. Curr. Protoc. Mouse Biol. 6 (2016), 333–342, 10.1002/cpmo.15.
Jellinger, K., et al. Brain iron and ferritin in Parkinson's and Alzheimer's diseases. J. Neural. Transm. Park. Dis. Dement. Sect. 2 (1990), 327–340, 10.1007/BF02252926.
Joppe, K., et al. The contribution of Iron to protein aggregation disorders in the central nervous system. Front. Neurosci., 13, 2019, 15, 10.3389/fnins.2019.00015.
Kaindlstorfer, C., et al. The relevance of Iron in the pathogenesis of multiple system atrophy: a viewpoint. J. Alzheimers Dis. 61 (2018), 1253–1273, 10.3233/JAD-170601.
Koskenkorva-Frank, T.S., et al. The complex interplay of iron metabolism, reactive oxygen species, and reactive nitrogen species: insights into the potential of various iron therapies to induce oxidative and nitrosative stress. Free Radic. Biol. Med. 65 (2013), 1174–1194, 10.1016/j.freeradbiomed.2013.09.001.
Kriks, S., et al. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature 480 (2011), 547–551, 10.1038/nature10648.
Kuhn, D.M., et al. Nitrotyrosine as a marker for peroxynitrite-induced neurotoxicity: the beginning or the end of the end of dopamine neurons?. J. Neurochem. 89 (2004), 529–536, 10.1111/j.1471-4159.2004.02346.x.
Lane, D.J., et al. Cellular iron uptake, trafficking and metabolism: key molecules and mechanisms and their roles in disease. Biochim. Biophys. Acta 1853 (2015), 1130–1144, 10.1016/j.bbamcr.2015.01.021.
Lang, M., et al. A genome on shaky ground: exploring the impact of mitochondrial DNA integrity on Parkinson's disease by highlighting the use of cybrid models. Cell. Mol. Life Sci., 79, 2022, 283, 10.1007/s00018-022-04304-3.
Lau, A., Tymianski, M., Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 460 (2010), 525–542, 10.1007/s00424-010-0809-1.
Lemieux, H., et al. Remodeling pathway control of mitochondrial respiratory capacity by temperature in mouse heart: electron flow through the Q-junction in permeabilized fibers. Sci. Rep., 7, 2017, 2840, 10.1038/s41598-017-02789-8.
Levin, J., et al. Elevated levels of methylmalonate and homocysteine in Parkinson's disease, progressive supranuclear palsy and amyotrophic lateral sclerosis. Dement. Geriatr. Cogn. Disord. 29 (2010), 553–559, 10.1159/000314841.
Lhermitte, J., et al. Original papers: on the occurrence of abnormal deposits of iron in the brain in parkinsonism with special reference to its localisation. J. Neurol. Psychopathol. 5 (1924), 195–208, 10.1136/jnnp.s1-5.19.195.
Li, S.W., et al. Iron overload induced by ferric ammonium citrate triggers reactive oxygen species-mediated apoptosis via both extrinsic and intrinsic pathways in human hepatic cells. Hum. Exp. Toxicol. 35 (2016), 598–607, 10.1177/0960327115597312.
Liang, L.P., Patel, M., Iron-sulfur enzyme mediated mitochondrial superoxide toxicity in experimental Parkinson's disease. J. Neurochem. 90 (2004), 1076–1084, 10.1111/j.1471-4159.2004.02567.x.
Lill, R., Freibert, S.A., Mechanisms of mitochondrial iron-sulfur protein biogenesis. Annu. Rev. Biochem. 89 (2020), 471–499, 10.1146/annurev-biochem-013118-111540.
Lin, T.K., et al. Mitochondrial dysfunction and biogenesis in the pathogenesis of Parkinson's disease. Chang Gung Med. J. 32 (2009), 589–599.
Liu, R.Z., et al. Baicalein attenuates brain iron accumulation through protecting aconitase 1 from oxidative stress in rotenone-induced Parkinson's disease in rats. Antioxidants (Basel), 12, 2022, 10.3390/antiox12010012.
Ma, L., et al. Parkinson's disease: alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol., 41, 2021, 101896, 10.1016/j.redox.2021.101896.
Malpartida, A.B., et al. Mitochondrial dysfunction and mitophagy in Parkinson's disease: from mechanism to therapy. Trends Biochem. Sci. 46 (2021), 329–343, 10.1016/j.tibs.2020.11.007.
Marras, C., et al. Nomenclature of genetic movement disorders: recommendations of the international Parkinson and movement disorder society task force. Mov. Disord. 31 (2016), 436–457, 10.1002/mds.26527.
Martin-Bastida, A., et al. Iron and inflammation: in vivo and post-mortem studies in Parkinson's disease. J. Neural Transm. (Vienna) 128 (2021), 15–25, 10.1007/s00702-020-02271-2.
Matak, P., et al. Disrupted iron homeostasis causes dopaminergic neurodegeneration in mice. Proc. Natl. Acad. Sci. U. S. A. 113 (2016), 3428–3435, 10.1073/pnas.1519473113.
Mercuri, N.B., et al. Tranylcypromine, but not moclobemide, prolongs the inhibitory action of dopamine on midbrain dopaminergic neurons: an in vitro electrophysiological study. Synapse 37 (2000), 216–221, 10.1002/1098-2396(20000901)37:3<216::AID-SYN5>3.0.CO;2-3.
Miyazaki, I., Asanuma, M., Dopaminergic neuron-specific oxidative stress caused by dopamine itself. Acta Med. Okayama 62 (2008), 141–150, 10.18926/AMO/30942.
Monzio Compagnoni, G., et al. The role of mitochondria in neurodegenerative diseases: the lesson from Alzheimer's disease and Parkinson's disease. Mol. Neurobiol. 57 (2020), 2959–2980, 10.1007/s12035-020-01926-1.
Muckenthaler, M.U., et al. A red carpet for iron metabolism. Cell 168 (2017), 344–361, 10.1016/j.cell.2016.12.034.
Muhlenhoff, U., et al. Compartmentalization of iron between mitochondria and the cytosol and its regulation. Eur. J. Cell Biol. 94 (2015), 292–308, 10.1016/j.ejcb.2015.05.003.
Munoz, Y., et al. Parkinson's disease: the mitochondria-iron link. Parkinsons Dis., 2016, 2016, 7049108, 10.1155/2016/7049108.
Nuzzo, T., et al. The levels of the NMDA receptor co-agonist D-serine are reduced in the substantia nigra of MPTP-lesioned macaques and in the cerebrospinal fluid of Parkinson's disease patients. Sci. Rep., 9, 2019, 8898, 10.1038/s41598-019-45419-1.
Oexle, H., et al. Iron-dependent changes in cellular energy metabolism: influence on citric acid cycle and oxidative phosphorylation. Biochim. Biophys. Acta 1413 (1999), 99–107, 10.1016/s0005-2728(99)00088-2.
Ortega, R., et al. Alpha-synuclein over-expression induces increased iron accumulation and redistribution in Iron-exposed neurons. Mol. Neurobiol. 53 (2016), 1925–1934, 10.1007/s12035-015-9146-x.
Pantopoulos, K., et al. Differences in the regulation of iron regulatory protein-1 (IRP-1) by extra- and intracellular oxidative stress. J. Biol. Chem. 272 (1997), 9802–9808, 10.1074/jbc.272.15.9802.
Parihar, M.S., et al. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell. Mol. Life Sci. 65 (2008), 1272–1284, 10.1007/s00018-008-7589-1.
Park, J.S., et al. Serum methylmalonic acid correlates with neuropathic pain in idiopathic Parkinson's disease. Neurol. Sci. 38 (2017), 1799–1804, 10.1007/s10072-017-3056-9.
Park, J.S., et al. Mitochondrial dysfunction in Parkinson's disease: new mechanistic insights and therapeutic perspectives. Curr. Neurol. Neurosci. Rep., 18, 2018, 21, 10.1007/s11910-018-0829-3.
Post, B., et al. Young onset Parkinson's disease: a modern and tailored approach. J. Parkinsons Dis. 10 (2020), S29–S36, 10.3233/JPD-202135.
Prasuhn, J., et al. Levodopa impairs the energy metabolism of the basal ganglia in vivo. Ann. Neurol., 2024, 10.1002/ana.26884.
Pringsheim, T., et al. The prevalence of Parkinson's disease: a systematic review and meta-analysis. Mov. Disord. 29 (2014), 1583–1590, 10.1002/mds.25945.
Qi, A., et al. Plasma metabolic analysis reveals the dysregulation of short-chain fatty acid metabolism in Parkinson's disease. Mol. Neurobiol. 60 (2023), 2619–2631, 10.1007/s12035-022-03157-y.
Riederer, P., et al. Neurochemical perspectives to the function of monoamine oxidase. Acta Neurol. Scand. Suppl. 126 (1989), 41–45, 10.1111/j.1600-0404.1989.tb01781.x.
Rouault, T.A., The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat. Chem. Biol. 2 (2006), 406–414, 10.1038/nchembio807.
Ruttkies, C., et al. MetFrag relaunched: incorporating strategies beyond in silico fragmentation. J. Chem., 8, 2016, 3, 10.1186/s13321-016-0115-9.
Schymanski, E.L., et al. Identifying small molecules via high resolution mass spectrometry: communicating confidence. Environ. Sci. Technol. 48 (2014), 2097–2098, 10.1021/es5002105.
Schymanski, E.L., et al. Empowering large chemical knowledge bases for exposomics: PubChemLite meets MetFrag. J. Chem., 13, 2021, 19, 10.1186/s13321-021-00489-0.
Serra, M., et al. Perturbation of serine enantiomers homeostasis in the striatum of MPTP-lesioned monkeys and mice reflects the extent of dopaminergic midbrain degeneration. Neurobiol. Dis., 184, 2023, 106226, 10.1016/j.nbd.2023.106226.
Shao, Y., Le, W., Recent advances and perspectives of metabolomics-based investigations in Parkinson's disease. Mol. Neurodegener., 14, 2019, 3, 10.1186/s13024-018-0304-2.
Shibasaki, Y., et al. High-resolution mapping of SNCA encoding alpha-synuclein, the non-A beta component of Alzheimer's disease amyloid precursor, to human chromosome 4q21.3–>q22 by fluorescence in situ hybridization. Cytogenet. Cell Genet. 71 (1995), 54–55, 10.1159/000134061.
Spillantini, M.G., et al. Alpha-synuclein in filamentous inclusions of Lewy bodies from Parkinson's disease and dementia with Lewy bodies. Proc. Natl. Acad. Sci. U. S. A. 95 (1998), 6469–6473, 10.1073/pnas.95.11.6469.
Sulzer, D., et al. Neuromelanin biosynthesis is driven by excess cytosolic catecholamines not accumulated by synaptic vesicles. Proc. Natl. Acad. Sci. U. S. A. 97 (2000), 11869–11874, 10.1073/pnas.97.22.11869.
Sun, Y., et al. Kinetic modeling of pH-dependent oxidation of dopamine by iron and its relevance to Parkinson's disease. Front. Neurosci., 12, 2018, 859, 10.3389/fnins.2018.00859.
Swerdlow, R.H., et al. Origin and functional consequences of the complex I defect in Parkinson's disease. Ann. Neurol. 40 (1996), 663–671, 10.1002/ana.410400417.
Talavera Andujar, B., et al. Studying the Parkinson's disease metabolome and exposome in biological samples through different analytical and cheminformatics approaches: a pilot study. Anal. Bioanal. Chem. 414 (2022), 7399–7419, 10.1007/s00216-022-04207-z.
Teppola, H., et al. Morphological differentiation towards neuronal phenotype of SH-SY5Y neuroblastoma cells by estradiol, retinoic acid and cholesterol. Neurochem. Res. 41 (2016), 731–747, 10.1007/s11064-015-1743-6.
Theurl, I., et al. On-demand erythrocyte disposal and iron recycling requires transient macrophages in the liver. Nat. Med. 22 (2016), 945–951, 10.1038/nm.4146.
Tian, Y., et al. FTH1 inhibits ferroptosis through ferritinophagy in the 6-OHDA model of Parkinson's disease. Neurotherapeutics 17 (2020), 1796–1812, 10.1007/s13311-020-00929-z.
Tsugawa, H., et al. A lipidome atlas in MS-DIAL 4. Nat. Biotechnol. 38 (2020), 1159–1163, 10.1038/s41587-020-0531-2.
Volani, C., et al. Dietary iron loading negatively affects liver mitochondrial function. Metallomics 9 (2017), 1634–1644, 10.1039/c7mt00177k.
Wang, X., et al. Antibiotic use and abuse: a threat to mitochondria and chloroplasts with impact on research, health, and environment. Bioessays 37 (2015), 1045–1053, 10.1002/bies.201500071.
Ward, R.J., et al. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 13 (2014), 1045–1060, 10.1016/S1474-4422(14)70117-6.
Wasner, K., Smajic, S., Ghelfi, J., Delcambre, S., Prada-Medina, C.A., Knappe, E., Arena, G., Mulica, P., Agyeah, G., Rakovic, A., Boussaad, I., Badanjak, K., Ohnmacht, J., Gérardy, J.J., Takanashi, M., Trinh, J., Mittelbronn, M., Hattori, N., Klein, C., Antony, P., Seibler, P., Spielmann, M., Pereira, S.L., Grünewald, A., Parkin Deficiency Impairs Mitochondrial DNA Dynamics and Propagates Inflammation. Mov Disord 37:7 (2022 Jul), 1405–1415, 10.1002/mds.29025.
Werner, E.R., et al. Tetrahydrobiopterin: biochemistry and pathophysiology. Biochem. J. 438 (2011), 397–414, 10.1042/BJ20110293.
Wongkittichote, P., et al. Tricarboxylic acid cycle enzyme activities in a mouse model of methylmalonic aciduria. Mol. Genet. Metab. 128 (2019), 444–451, 10.1016/j.ymgme.2019.10.007.
Yan, M.H., et al. Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic. Biol. Med. 62 (2013), 90–101, 10.1016/j.freeradbiomed.2012.11.014.
Zambon, F., et al. Cellular alpha-synuclein pathology is associated with bioenergetic dysfunction in Parkinson's iPSC-derived dopamine neurons. Hum. Mol. Genet. 28 (2019), 2001–2013, 10.1093/hmg/ddz038.
Zanon, A., et al. SLP-2 interacts with Parkin in mitochondria and prevents mitochondrial dysfunction in Parkin-deficient human iPSC-derived neurons and Drosophila. Hum. Mol. Genet. 26 (2017), 2412–2425, 10.1093/hmg/ddx132.
Zhang, Z., et al. Roles of glutamate receptors in Parkinson's disease. Int. J. Mol. Sci., 20, 2019, 10.3390/ijms20184391.
Zhang, Y., et al. Iron acquisition by bacterial pathogens: beyond tris-catecholate complexes. Chembiochem 21 (2020), 1955–1967, 10.1002/cbic.201900778.
Zhou, Z.D., Tan, E.K., Iron regulatory protein (IRP)-iron responsive element (IRE) signaling pathway in human neurodegenerative diseases. Mol. Neurodegener., 12, 2017, 75, 10.1186/s13024-017-0218-4.
Zucca, F.A., et al. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson's disease. Prog. Neurobiol. 155 (2017), 96–119, 10.1016/j.pneurobio.2015.09.012.