Submitted:
21 January 2025
Posted:
22 January 2025
You are already at the latest version
Abstract
I present a complete theory of Parkinson’s disease (PD). PD involves an abundance of gut microbiota that degrade mucin, resulting in elevated brain uptake of galactose (a mucin component) and reduced uptake of glucose. Since galactose is not efficiently used for glycolysis, this decreases the production of glutathione, which is essential for countering oxidative iron toxicity. The brain cells that are most sensitive to this effect, as attested by their expression of the iron chelator neuromelanin, are those that continuously secrete dopamine and norepinephrine, and they eventually degenerate. This account explains the etiology, symptoms (including prodromal autonomic symptoms), pathology and risk factors of the disease. The theory is supported by strong evidence, and gives rise to several relatively simply directions for early identification and treatment of the disease.
Keywords:
1. Introduction
2. T*PD Theory of PD
- PD involves an imbalance between galactose and glucose in favor of the former. This yields reduced glycolysis.
- The main source for this imbalance is a higher abundance of mucin-degrading bacteria in the gut microbiome (GMB).
- The imbalance yields GSH deficiency, exposing cells to oxidative iron toxicity. The cells that are most sensitive to this are those that continuously secrete catecholamines and express neuromelanin.
- The phenomenon is a small-scale one, and the main damage occurs with the deterioration of cellular defenses with aging, and with the accumulation of NM and alpha-synuclein (aSyn).
- Some genetic mutations can enhance these processes.
3. Evidence
3.1. Gut Bacteria, Mucin
3.2. Galactose
3.3. Glucose
3.4. Oxidative Stress
3.5. Sympathetic Nervous System
3.6. Risk & Protective Factors
4. Treatment
5. Discussion
Acknowledgments
Abbreviations
| akkm | Akkermansia Muciniphila. |
| aSyn | alpha-synuclein. |
| GMB | gut microbiome. |
| GSH | glutathione. |
| HPL | helicobacter pylori. |
| LC | locus coeruelus. |
| NEP | norepinephrine. |
| NM | neuromelanin. |
| PD | Parkinson’s disease. |
| PPP | pentose phosphate pathway. |
| RBD | REM sleep behavior disorder. |
| SNc | substantia nigra pars compacta. |
| SNS | sympathetic nervous system. |
| TBI | traumatic brain injury. |
| T*PD | the theory of PD presented in this paper. |
References
- Obeso J, Goetz C, Lang A, Weintraub D, Bezard E, et al. Past, present, and future of Parkinson’s disease: A special essay on the 200th Anniversary of the Shaking Palsy. Movement disorders. 2017;32(9):1264-310.
- Morris HR, Spillantini MG, Sue CM, Williams-Gray CH. The pathogenesis of Parkinson’s disease. The Lancet. 2024;403(10423):293-304. [CrossRef]
- Surmeier DJ, Obeso JA, Halliday GM. Selective neuronal vulnerability in Parkinson disease. Nat Rev Neurosci. 2017;18(2):101. [CrossRef]
- Sara, SJ. The locus coeruleus and noradrenergic modulation of cognition. Nature reviews neuroscience. 2009;10(3):211-23. [CrossRef]
- Adams-Carr KL, Bestwick JP, Shribman S, Lees A, Schrag A, Noyce AJ. Constipation preceding Parkinson’s disease: a systematic review and meta-analysis. Journal of Neurology, Neurosurgery & Psychiatry. 2016;87(7):710-6.
- Guiney SJ, Adlard PA, Bush AI, Finkelstein DI, Ayton S. Ferroptosis and cell death mechanisms in Parkinson’s disease. Neurochemistry international. 2017;104:34-48.
- Zucca FA, Segura-Aguilar J, Ferrari E, Muñoz P, Paris I, Sulzer D, et al. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson’s disease. Progress in neurobiology. 2017;155:96-119.
- Jansen van Rensburg Z, Abrahams S, Bardien S, Kenyon C. Toxic feedback loop involving iron, reactive oxygen species, α-synuclein and neuromelanin in Parkinson’s disease and intervention with turmeric. Molecular Neurobiology. 2021;58(11):5920-36.
- Conte F, van Buuringen N, Voermans NC, Lefeber DJ. Galactose in human metabolism, glycosylation and congenital metabolic diseases: Time for a closer look. Biochimica et Biophysica Acta (BBA)-General Subjects. 2021;1865(8):129898.
- Roser M, Josic D, Kontou M, Mosetter K, Maurer P, Reutter W. Metabolism of galactose in the brain and liver of rats and its conversion into glutamate and other amino acids. Journal of neural transmission. 2009;116:131-9.
- Henderson JM, Kutner MH, Bain RP. First-order clearance of plasma galactose: the effect of liver disease. Gastroenterology. 1982;83(5):1090-6.
- Aguer C, Gambarotta D, Mailloux RJ, Moffat C, Dent R, McPherson R, et al. Galactose enhances oxidative metabolism and reveals mitochondrial dysfunction in human primary muscle cells. PloS one. 2011;6(12):e28536.
- Shwe T, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Role of D-galactose-induced brain aging and its potential used for therapeutic interventions. Experimental gerontology. 2018;101:13-36.
- Rossignol R, Gilkerson R, Aggeler R, Yamagata K, Remington SJ, Capaldi RA. Energy substrate modulates mitochondrial structure and oxidative capacity in cancer cells. Cancer research. 2004;64(3):985-93.
- Cui X, Zuo P, Zhang Q, Li X, Hu Y, Long J, et al. Chronic systemic D-galactose exposure induces memory loss, neurodegeneration, and oxidative damage in mice: Protective effects of R-α-lipoic acid. Journal of neuroscience research. 2006;83(8):1584-90.
- Xia MY, Zhao XY, Huang QL, Sun HY, Sun C, Yuan J, et al. Activation of Wnt/β-catenin signaling by lithium chloride attenuates d-galactose-induced neurodegeneration in the auditory cortex of a rat model of aging. FEBS open bio. 2017;7(6):759-76.
- Gibbs ME, Hutchinson DS, Summers RJ. Role of β-adrenoceptors in memory consolidation: β3-adrenoceptors act on glucose uptake and β2-adrenoceptors on glycogenolysis. Neuropsychopharmacology. 2008;33(10):2384-97.
- Brockhausen I, Schachter H, Stanley P. O-GalNAc glycans. Essentials of Glycobiology 2nd edition. 2009.
- Yamaguchi M, Yamamoto K. Mucin glycans and their degradation by gut microbiota. Glycoconjugate Journal. 2023;40(4):493-512.
- Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. International journal of systematic and evolutionary microbiology. 2004;54(5):1469-76.
- Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proceedings of the national academy of sciences. 2013;110(22):9066-71.
- Martin-Gallausiaux C, Garcia-Weber D, Lashermes A, Larraufie P, Marinelli L, Teixeira V, et al. Akkermansia muciniphila upregulates genes involved in maintaining the intestinal barrier function via ADP-heptose-dependent activation of the ALPK1/TIFA pathway. Gut microbes. 2022;14(1):2110639.
- Depommier C, Van Hul M, Everard A, Delzenne NM, De Vos WM, Cani PD. Pasteurized Akkermansia muciniphila increases whole-body energy expenditure and fecal energy excretion in diet-induced obese mice. Gut microbes. 2020;11(5):1231-45.
- Xu Y, Yang Y, Li B, Xie Y, Shi Y, Le G. Dietary methionine restriction improves gut microbiota composition and prevents cognitive impairment in D-galactose-induced aging mice. Food & Function. 2022;13(24):12896-914.
- Kim JJ, Khan WI. Goblet cells and mucins: role in innate defense in enteric infections. pathogens. 2013;2(1):55-70.
- Chen L, Collij V, Hu S, et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature. 2022;604(7907):732-9.
- Pacelli C, Giguère N, Bourque MJ, Lévesque M, Slack RS, Trudeau LÉ. Elevated mitochondrial bioenergetics and axonal arborization size are key contributors to the vulnerability of dopamine neurons. Current Biology. 2015;25(18):2349-60.
- Stincone A, Prigione A, Cramer T, Wamelink MM, Campbell K, Cheung E, et al. The return of metabolism: biochemistry and physiology of the pentose phosphate pathway. Biological Reviews. 2015;90(3):927-63.
- Zecca L, Casella L, Albertini A, Bellei C, Zucca FA, Engelen M, et al. Neuromelanin can protect against iron-mediated oxidative damage in system modeling iron overload of brain aging and Parkinson’s disease. Journal of neurochemistry. 2008;106(4):1866-75.
- Bloem BR, Okun MS, Klein C. Parkinson’s disease. The Lancet. 2021;397(10291):2284-303.
- Mattson MP, Arumugam TV. Hallmarks of brain aging: adaptive and pathological modification by metabolic states. Cell metabolism. 2018;27(6):1176-99.
- Schnegg M, Lauterburg BH. Quantitative liver function in the elderly assessed by galactose elimination capacity, aminopyrine demethylation and caffeine clearance. Journal of hepatology. 1986;3(2):164-71.
- Suh JH, Shenvi SV, Dixon BM, Liu H, Jaiswal AK, Liu RM, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proceedings of the National Academy of Sciences. 2004;101(10):3381-6.
- Hebbrecht G, Maenhaut W, De Reuck J. Brain trace elements and aging. Nuclear instruments and methods in physics research section B: beam interactions with materials and atoms. 1999;150(1-4):208-13.
- Floor E, Wetzel MG. Increased protein oxidation in human substantia nigra pars compacta in comparison with basal ganglia and prefrontal cortex measured with an improved dinitrophenylhydrazine assay. Journal of neurochemistry. 1998;70(1):268-75.
- Hashimoto M, Takeda A, Hsu LJ, Takenouchi T, Masliah E. Role of cytochrome c as a stimulator of α-synuclein aggregation in Lewy body disease. Journal of Biological Chemistry. 1999;274(41):28849-52.
- Chen B, Wen X, Jiang H, Wang J, Song N, Xie J. Interactions between iron and α-synuclein pathology in Parkinson’s disease. Free Radical Biology and Medicine. 2019;141:253-60.
- Xuan Q, Xu SL, Lu DH, Yu S, Zhou M, Uéda K, et al. Increase expression of α-synuclein in aged human brain associated with neuromelanin accumulation. Journal of neural transmission. 2011;118:1575-83.
- Zhu M, Qin ZJ, Hu D, Munishkina LA, Fink AL. α-synuclein can function as an antioxidant preventing oxidation of unsaturated lipid in vesicles. Biochemistry. 2006;45(26):8135-42. [CrossRef]
- Musgrove RE, King AE, Dickson TC. α-Synuclein protects neurons from apoptosis downstream of free-radical production through modulation of the MAPK signalling pathway. Neurotoxicity research. 2013;23:358-69. [CrossRef]
- Mizuno N, Varkey J, Kegulian NC, Hegde BG, Cheng N, Langen R, et al. Remodeling of lipid vesicles into cylindrical micelles by α-synuclein in an extended α-helical conformation. Journal of Biological Chemistry. 2012;287(35):29301-11.
- Kamp F, Exner N, Lutz AK, Wender N, Hegermann J, Brunner B, et al. Inhibition of mitochondrial fusion by α-synuclein is rescued by PINK1, Parkin and DJ-1. The EMBO journal. 2010;29(20):3571-89.
- Paillusson S, Gomez-Suaga P, Stoica R, Little D, Gissen P, Devine MJ, et al. α-Synuclein binds to the ER–mitochondria tethering protein VAPB to disrupt Ca2+ homeostasis and mitochondrial ATP production. Acta neuropathologica. 2017;134:129-49.
- Rodriguez-Araujo G, Nakagami H, Hayashi H, Mori M, Shiuchi T, Minokoshi Y, et al. Alpha-synuclein elicits glucose uptake and utilization in adipocytes through the Gab1/PI3K/Akt transduction pathway. Cellular and molecular life sciences. 2013;70:1123-33.
- Qiao H, He X, Zhang Q, Yuan H, Wang D, Li L, et al. Alpha-synuclein induces microglial migration via PKM2-dependent glycolysis. International journal of biological macromolecules. 2019;129:601-7.
- Chau KY, Ching HL, Schapira AH, Cooper JM. Relationship between alpha synuclein phosphorylation, proteasomal inhibition and cell death: relevance to Parkinson’s disease pathogenesis. Journal of neurochemistry. 2009;110(3):1005-13.
- Dias V, Junn E, Mouradian MM. The role of oxidative stress in Parkinson’s disease. Journal of Parkinson’s disease. 2013;3(4):461-91.
- Gegg ME, Schapira AH. The role of glucocerebrosidase in Parkinson disease pathogenesis. The FEBS journal. 2018;285(19):3591-603.
- Hill-Burns EM, Debelius JW, Morton JT, Wissemann WT, Lewis MR, Wallen ZD, et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Movement disorders. 2017;32(5):739-49.
- Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Parkinson’s Disease. 2021;7(1):27.
- Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Movement Disorders. 2015;30(3):350-8.
- Murakami K, Okubo H, Sasaki S. Dietary intake in relation to self-reported constipation among Japanese women aged 18–20 years. European journal of clinical nutrition. 2006;60(5):650-7.
- Adolf LA, Heilbronner S. Nutritional interactions between bacterial species colonising the human nasal cavity: current knowledge and future prospects. Metabolites. 2022;12(6):489.
- Ma LY, Liu GL, Wang DX, Zhang MM, Kou WY, Feng T. Alpha-synuclein in peripheral tissues in Parkinson’s disease. ACS Chemical Neuroscience. 2019;10(2):812-23.
- Wang H, Liu X, Tan C, Zhou W, Jiang J, Peng W, et al. Bacterial, viral, and fungal infection-related risk of Parkinson’s disease: Meta-analysis of cohort and case–control studies. Brain and behavior. 2020;10(3):e01549.
- Morgante L, Salemi G, Meneghini F, Di Rosa AE, Epifanio A, Grigoletto F, et al. Parkinson disease survival: a population-based study. Archives of neurology. 2000;57(4):507-12.
- Ryu DW, Han K, Cho AH. Mortality and causes of death in patients with Parkinson’s disease: a nationwide population-based cohort study. Frontiers in Neurology. 2023;14:1236296.
- Slomiany B, Murty V, Piotrowski J, Liau Y, Sundaram P, Slomiany A. Glycosulfatase activity of Helicobacter pylori toward gastric mucin. Biochemical and biophysical research communications. 1992;183(2):506-13.
- Murata H, Barnhill LM, Bronstein JM. Air pollution and the risk of Parkinson’s disease: a review. Movement Disorders. 2022;37(5):894-904.
- Gillois K, Lévêque M, Théodorou V, Robert H, Mercier-Bonin M. Mucus: an underestimated gut target for environmental pollutants and food additives. Microorganisms. 2018;6(2):53.
- Ascherio A, Schwarzschild MA. The epidemiology of Parkinson’s disease: risk factors and prevention. The Lancet Neurology. 2016;15(12):1257-72.
- Luna E, Parkar SG, Kirmiz N, Hartel S, Hearn E, Hossine M, et al. Utilization efficiency of human milk oligosaccharides by human-associated Akkermansia is strain dependent. Applied and Environmental Microbiology. 2022;88(1):e01487-21.
- Wuolikainen A, Jonsson P, Ahnlund M, Antti H, Marklund SL, Moritz T, et al. Multi-platform mass spectrometry analysis of the CSF and plasma metabolomes of rigorously matched amyotrophic lateral sclerosis, Parkinson’s disease and control subjects. Molecular BioSystems. 2016;12(4):1287-98.
- Scholefield M, Church SJ, Taylor G, Knight D, Unwin RD, Cooper GJ. Multi-regional alterations in glucose and purine metabolic pathways in the Parkinson’s disease dementia brain. npj Parkinson’s Disease. 2023;9(1):66.
- Ahmed SS, Santosh W, Kumar S, Christlet HTT. Metabolic profiling of Parkinson’s disease: evidence of biomarker from gene expression analysis and rapid neural network detection. Journal of biomedical science. 2009;16(1):1-12.
- Konjevod M, Sáiz J, Barbas C, Bergareche A, Ardanaz E, Huerta JM, et al. A Set of Reliable Samples for the Study of Biomarkers for the Early Diagnosis of Parkinson’s Disease. Frontiers in Neurology. 2022;13:844841.
- Videira PA, Castro-Caldas M. Linking glycation and glycosylation with inflammation and mitochondrial dysfunction in Parkinson’s disease. Frontiers in neuroscience. 2018;12:381.
- Panis B, Bosch AM, Burlina A, Coman DJ, Couce ML, Das AM, et al. Brain function in classic galactosemia, a galactosemia network (GalNet) members review. Frontiers in Genetics. 2024;15:1355962.
- Rubio-Agusti I, Carecchio M, Bhatia KP, Kojovic M, Parees I, Chandrashekar HS, et al. Movement disorders in adult patients with classical galactosemia. Movement Disorders. 2013;28(6):804-10.
- Poisson A, Roze E, Demily C, Thobois S. Evidence for dopaminergic denervation in classical galactosemia. Movement disorders: official journal of the Movement Disorder Society. 2017;32(6):940-2.
- Boca M, Whone A. Letter to the editor on “Evidence for dopaminergic denervation in classical galactosemia”. Movement Disorders: Official Journal of the Movement Disorder Society. 2017;32(12):1797-7.
- van Erven B, Römers MM, Rubio-Gozalbo ME. Revised proposal for the prevention of low bone mass in patients with classic galactosemia. In: JIMD Reports, Volume 17. Springer; 2014. p. 41-6.
- Torsney KM, Noyce AJ, Doherty KM, Bestwick JP, Dobson R, Lees AJ. Bone health in Parkinson’s disease: a systematic review and meta-analysis. Journal of Neurology, Neurosurgery & Psychiatry. 2014;85(10):1159-66.
- LeWitt PA, Li J, Lu M, Beach TG, Adler CH, Guo L, et al. 3-hydroxykynurenine and other Parkinson’s disease biomarkers discovered by metabolomic analysis. Movement Disorders. 2013;28(12):1653-60.
- Öhman A, Forsgren L. NMR metabonomics of cerebrospinal fluid distinguishes between Parkinson’s disease and controls. Neuroscience letters. 2015;594:36-9.
- Murakami K, Miyake Y, Sasaki S, Tanaka K, Fukushima W, Kiyohara C, et al. Dietary glycemic index is inversely associated with the risk of Parkinson’s disease: a case–control study in Japan. Nutrition. 2010;26(5):515-21.
- Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiology of aging. 2014;35(5):1111-5.
- Elstner M, Morris CM, Heim K, Bender A, Mehta D, Jaros E, et al. Expression analysis of dopaminergic neurons in Parkinson’s disease and aging links transcriptional dysregulation of energy metabolism to cell death. Acta neuropathologica. 2011;122:75-86.
- Ostrakhovitch EA, Song ES, Macedo JK, Gentry MS, Quintero JE, van Horne C, et al. Analysis of circulating metabolites to differentiate Parkinson’s disease and essential tremor. Neuroscience letters. 2022;769:136428.
- Nagao M, Hayashi H. Glycogen synthase kinase-3beta is associated with Parkinson’s disease. Neuroscience Letters. 2009;449(2):103-7.
- Rango M, Bonifati C, Bresolin N. Parkinson’s disease and brain mitochondrial dysfunction: a functional phosphorus magnetic resonance spectroscopy study. Journal of Cerebral Blood Flow & Metabolism. 2006;26(2):283-90.
- Simmering JE, Welsh MJ, Liu L, Narayanan NS, Pottegård A. Association of glycolysis-enhancing α-1 blockers with risk of developing Parkinson disease. JAMA neurology. 2021;78(4):407-13.
- Eberling JL, Richardson BC, Reed BR, Wolfe N, Jagust WJ. Cortical glucose metabolism in Parkinson’s disease without dementia. Neurobiology of Aging. 1994;15(3):329-35.
- Hosokai Y, Nishio Y, Hirayama K, Takeda A, Ishioka T, Sawada Y, et al. Distinct patterns of regional cerebral glucose metabolism in Parkinson’s disease with and without mild cognitive impairment. Movement disorders: official journal of the Movement Disorder Society. 2009;24(6):854-62.
- Xu Y, Wei X, Liu X, Liao J, Lin J, Zhu C, et al. Low cerebral glucose metabolism: a potential predictor for the severity of vascular parkinsonism and Parkinson’s disease. Aging and disease. 2015;6(6):426.
- Baba T, Takeda A, Kikuchi A, Nishio Y, Hosokai Y, Hirayama K, et al. Association of olfactory dysfunction and brain metabolism in Parkinson’s disease. Movement Disorders. 2011;26(4):621-8.
- Ådén E, Carlsson M, Poortvliet E, Stenlund H, Linder J, Edström M, et al. Dietary intake and olfactory function in patients with newly diagnosed Parkinson’s disease: a case-control study. Nutritional neuroscience. 2011;14(1):25-31.
- Hellenbrand W, Seidler A, Boeing H, Robra BP, Vieregge P, Nischan P, et al. Diet and Parkinson’s disease I: A possible role for the past intake of specific foods and food groups: Results from a self-administered food-frequency questionnaire in a case-control study. Neurology. 1996;47(3):636-43.
- Cassani E, Barichella M, Ferri V, Pinelli G, Iorio L, Bolliri C, et al. Dietary habits in Parkinson’s disease: adherence to Mediterranean diet. Parkinsonism & related disorders. 2017;42:40-6.
- Camargo Maluf F, Feder D, Alves de Siqueira Carvalho A, et al. Analysis of the relationship between type II diabetes mellitus and Parkinson’s disease: a systematic review. Parkinson’s Disease;2019.
- Sheard JM, Ash S, Silburn PA, Kerr GK. Prevalence of malnutrition in Parkinson’s disease: a systematic review. Nutrition reviews. 2011;69(9):520-32.
- Cheshire Jr WP, Wszolek ZK. Body mass index is reduced early in Parkinson’s disease. Parkinsonism & related disorders. 2005;11(1):35-8.
- Deus CM, Pereira SP, Cunha-Oliveira T, Pereira FB, Raimundo N, Oliveira PJ. Mitochondrial remodeling in human skin fibroblasts from sporadic male Parkinson’s disease patients uncovers metabolic and mitochondrial bioenergetic defects. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease. 2020;1866(3):165615.
- Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nature neuroscience. 2000;3(12):1301-6.
- Sian J, Dexter DT, Lees AJ, Daniel S, Agid Y, Javoy-Agid F, et al. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society. 1994;36(3):348-55.
- Kish SJ, Morito C, Hornykiewicz O. Glutathione peroxidase activity in Parkinson’s disease brain. Neuroscience letters. 1985;58(3):343-6.
- Sofic E, Jellinger K. Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson’s disease. Neuroscience letters. 1992;142(2):128-30.
- Jenner P, Dexter D, Sian J, Schapira A, Marsden C. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental Lewy body disease. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society. 1992;32(S1):S82-7.
- Pearce R, Owen A, Daniel S, Jenner P, Marsden C. Alterations in the distribution of glutathione in the substantia nigra in Parkinson’s disease. Journal of neural transmission. 1997;104:661-77.
- Riederer P, Sofic E, Rausch WD, Schmidt B, Reynolds GP, Jellinger K, et al. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. Journal of neurochemistry. 1989;52(2):515-20.
- Maetzler W, Schmid SP, Wurster I, Liepelt I, Gaenslen A, Gasser T, et al. Reduced but not oxidized cerebrospinal fluid glutathione levels are lowered in Lewy body diseases. Movement Disorders. 2011;26(1):176-81.
- Cristalli DO, Arnal N, Marra FA, de Alaniz MJ, Marra CA. Peripheral markers in neurodegenerative patients and their first-degree relatives. Journal of the neurological sciences. 2012;314(1-2):48-56.
- Charisis S, Ntanasi E, Stamelou M, Xiromerisiou G, Maraki M, Veskoukis AS, et al. Plasma glutathione and prodromal Parkinson’s disease probability. Movement Disorders. 2022;37(1):200-5.
- He Q, Zhang Z, Fu B, Chen J, Liu J. Changes in serum uric acid, glutathione, and amyloid-β1-42 levels in Parkinson’s disease patients and their association with disease progression and cognitive decline. Current Medical Research and Opinion. 2024;(just-accepted):1-15.
- Sofic E, Riederer P, Heinsen H, Beckmann H, Reynolds G, Hebenstreit G, et al. Increased iron (III) and total iron content in post mortem substantia nigra of parkinsonian brain. Journal of neural transmission. 1988;74:199-205.
- Dexter D, Wells F, Lee A, Agid F, Agid Y, Jenner P, et al. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson’s disease. Journal of neurochemistry. 1989;52(6):1830-6.
- Popescu BFG, George MJ, Bergmann U, Garachtchenko AV, Kelly ME, McCrea RP, et al. Mapping metals in Parkinson’s and normal brain using rapid-scanning x-ray fluorescence. Physics in Medicine & Biology. 2009;54(3):651.
- Martin WW, Wieler M, Gee M. Midbrain iron content in early Parkinson disease: a potential biomarker of disease status. Neurology. 2008;70(16 Part 2):1411-7.
- Yoritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER, Mizuno Y. Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. In: Proceedings of the National Academy of Science. vol. 93; 1996. p. 2696-701.
- Selley, ML. (E)-4-hydroxy-2-nonenal may be involved in the pathogenesis of Parkinson’s disease. Free Radical Biology and Medicine. 1998;25(2):169-74.
- Castellani RJ, Perry G, Siedlak SL, Nunomura A, Shimohama S, Zhang J, et al. Hydroxynonenal adducts indicate a role for lipid peroxidation in neocortical and brainstem Lewy bodies in humans. Neuroscience letters. 2002;319(1):25-8.
- Dalfó E, Ferrer I. Early α-synuclein lipoxidation in neocortex in Lewy body diseases. Neurobiology of Aging. 2008;29(3):408-17.
- Depierreux F, Parmentier E, Mackels L, Baquero K, Degueldre C, Balteau E, et al. Parkinson’s disease multimodal imaging: F-DOPA PET, neuromelanin-sensitive and quantitative iron-sensitive MRI. npj Parkinson’s Disease. 2021;7(1):57.
- Berg D, Postuma RB, Adler CH, Bloem BR, Chan P, Dubois B, et al. MDS research criteria for prodromal Parkinson’s disease. Movement Disorders. 2015;30(12):1600-11.
- Sääksjärvi K, Knekt P, Männistö S, Lyytinen J, Heliövaara M. Prospective study on the components of metabolic syndrome and the incidence of Parkinson’s disease. Parkinsonism & related disorders. 2015;21(10):1148-55.
- Trupp M, Jonsson P, Öhrfelt A, Zetterberg H, Obudulu O, Malm L, et al. Metabolite and peptide levels in plasma and CSF differentiating healthy controls from patients with newly diagnosed Parkinson’s disease. Journal of Parkinson’s disease. 2014;4(3):549-60.
- García-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, Leu-Semenescu S, Gallea C, Quattrocchi G, et al. The coeruleus/subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson’s disease. Brain. 2013;136(7):2120-9.
- Zhang D, Jiang H, Xie J. Alcohol intake and risk of Parkinson’s disease: a meta-analysis of observational studies. Movement Disorders. 2014;29(6):819-22.
- Capurso G, Lahner E. The interaction between smoking, alcohol and the gut microbiome. Best practice & research Clinical gastroenterology. 2017;31(5):579-88.
- Breckenridge CB, Berry C, Chang ET, Sielken Jr RL, Mandel JS. Association between Parkinson’s disease and cigarette smoking, rural living, well-water consumption, farming and pesticide use: systematic review and meta-analysis. PloS one. 2016;11(4):e0151841.
- Ullah F, Ali T, Ullah N, Kim MO. Caffeine prevents d-galactose-induced cognitive deficits, oxidative stress, neuroinflammation and neurodegeneration in the adult rat brain. Neurochemistry international. 2015;90:114-24.
- Xie BS, Wang YQ, Lin Y, Mao Q, Feng JF, Gao GY, et al. Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS neuroscience & therapeutics. 2019;25(4):465-75.
- Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson’s disease: systematic review and meta-analysis of observational studies. Drugs & aging. 2009;26:769-79.
- Paul KC, Chuang YH, Shih IF, Keener A, Bordelon Y, Bronstein JM, et al. The association between lifestyle factors and Parkinson’s disease progression and mortality. Movement Disorders. 2019;34(1):58-66.
- Bajaj A, Driver JA, Schernhammer ES. Parkinson’s disease and cancer risk: a systematic review and meta-analysis. Cancer Causes & Control. 2010;21:697-707.
- Bose A, Petsko GA, Eliezer D. Parkinson’s disease and melanoma: co-occurrence and mechanisms. Journal of Parkinson’s disease. 2018;8(3):385-98.
- Gronich N, Abernethy DR, Auriel E, Lavi I, Rennert G, Saliba W. β2-adrenoceptor agonists and antagonists and risk of Parkinson’s disease. Movement Disorders. 2018;33(9):1465-71. [CrossRef]
- Amano S, Kegelmeyer D, Hong SL. Rethinking energy in parkinsonian motor symptoms: a potential role for neural metabolic deficits. Frontiers in systems neuroscience. 2015;8:242.
- Sarni AR, Baroni L. Milk and Parkinson disease: Could galactose be the missing link. Mediterranean Journal of Nutrition and Metabolism. 2019;12(1):91-118.
- Adams, JD. Possible causes of Parkinson’s disease. Frontiers in Bioscience-Landmark. 2021;26(8):387-94. [CrossRef]
- Munoz-Pinto MF, Empadinhas N, Cardoso SM. The neuromicrobiology of Parkinson’s disease: a unifying theory. Ageing Research Reviews. 2021;70:101396.
- Engelender S, Isacson O. The threshold theory for Parkinson’s disease. Trends in neurosciences. 2017;40(1):4-14.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).