Document Type : Review Article

Authors

1 Faculty of Pharmacy and Biochemistry, National University of San Marcos, Lima 03, Peru

2 Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

3 Faculty of Pharmacy, Pontifical Catholic University of Peru, Lima 03, Peru

Abstract

It is possible for the non-motor symptoms (NMS) of Parkinson's disease (PD), which include constipation, sleep difficulties, and olfactory impairments, to appear up to 20 years before the motor symptoms of the disease. There is a growing body of research that suggests the pathology of Parkinson's disease may begin in the gastrointestinal tract and progress to the brain. Numerous studies provide credence to the idea that the microbiota in one's gut communicates with one's brain in Parkinson's disease (PD) via way of the immune system, a certain amino acid metabolism, and the neurological system. Through what has become known as the "gut microbiota-brain axis" (GMBA), the gut microbiota is thought to play an important part in the modulation of several neurochemical pathways.In the process of mediating the crosstalk between the gut microbiota and the physiology of the host, many of the metabolites produced by the gut microbiota, such as fatty acids, amino acids, and bile acids, carry signaling activities. In Parkinson's disease (PD), the quantity of amino acids and species-specific alterations of amino acids, such as glutamate and tryptophan, may interfere with the signaling transmission between nerve cells and disrupt the normal operation of the basal ganglia. Certain amino acids and the receptors that bind to them are being looked at as new possible targets for the treatment of PD. The purpose of the current investigation was to compile and analyze all of the evidence that is currently available on the gut microbiota-derived amino acid metabolic changes that are related with PD.

Graphical Abstract

Gut microbiota and parkinson's disease

Keywords

Main Subjects

Selected author of this article by journal

ِDr. Mohammad Reza Mohammadi
Tarbiat Modares University

Google Scholar

Open Access

This article is licensed under a CC BY License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit: http://creativecommons.org/licenses/by/4.0/

 

Publisher’s Note

CMBR journal remains neutral with regard to jurisdictional claims in published maps and institutional afflictions.

 

Letters to Editor

Given that CMBR Journal's policy in accepting articles will be strict and will do its best to ensure that in addition to having the highest quality published articles, the published articles should have the least similarity (maximum 15%). Also, all the figures and tables in the article must be original and the copyright permission of images must be prepared by authors. However, some articles may have flaws and have passed the journal filter, which dear authors may find fault with. Therefore, the editor of the journal asks the authors, if they see an error in the published articles of the journal, to email the article information along with the documents to the journal office.

CMBR Journal welcomes letters to the editor ([email protected], [email protected]) for the post-publication discussions and corrections which allows debate post publication on its site, through the Letters to Editor. Critical letters can be sent to the journal editor as soon as the article is online. Following points are to be considering before sending the letters (comments) to the editor.

[1] Letters that include statements of statistics, facts, research, or theories should include appropriate references, although more than three are discouraged.

[2] Letters that are personal attacks on an author rather than thoughtful criticism of the author’s ideas will not be considered for publication.

[3] There is no limit to the number of words in a letter.

[4] Letter writers should include a statement at the beginning of the letter stating that it is being submitted either for publication or not.

[5] Anonymous letters will not be considered.

[6] Letter writers must include Name, Email Address, Affiliation, mobile phone number, and Comments.

[7] Letters will be answered as soon as possible.

  1. Goralczyk-Binkowska A, Szmajda-Krygier D, Kozlowska E (2022) The Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int J Mol Sci 23 (19). doi: https://doi.org/10.3390/ijms231911245
  2. Li C, Liang Y, Qiao Y (2022) Messengers From the Gut: Gut Microbiota-Derived Metabolites on Host Regulation. Front Microbiol 13: 863407. doi: https://doi.org/10.3389/fmicb.2022.863407
  3. Ortega MA, Alvarez-Mon MA, Garcia-Montero C, Fraile-Martinez O, Guijarro LG, Lahera G, Monserrat J, Valls P, Mora F, Rodriguez-Jimenez R, Quintero J, Alvarez-Mon M (2022) Gut Microbiota Metabolites in Major Depressive Disorder-Deep Insights into Their Pathophysiological Role and Potential Translational Applications. Metabolites 12 (1). doi: https://doi.org/10.3390/metabo12010050
  4. Pinheiro Campos AC, Martinez RCR, Auada AVV, Lebrun I, Fonoff ET, Hamani C, Pagano RL (2022) Effect of Subthalamic Stimulation and Electrode Implantation in the Striatal Microenvironment in a Parkinson's Disease Rat Model. Int J Mol Sci 23 (20). doi: https://doi.org/10.3390/ijms232012116
  5. Sun Y, Wang S, Liu B, Hu W, Zhu Y (2023) Host-Microbiome Interactions: Tryptophan Metabolism and Aromatic Hydrocarbon Receptors after Traumatic Brain Injury. Int J Mol Sci 24 (13). doi: https://doi.org/10.3390/ijms241310820
  6. Tamura H, Nishio R, Saeki N, Katahira M, Morioka H, Tamano H, Takeda A (2022) Paraquat-induced intracellular Zn(2+) dysregulation causes dopaminergic degeneration in the substantia nigra, but not in the striatum. Neurotoxicology 90: 136-144. doi: https://doi.org/10.1016/j.neuro.2022.03.010
  7. Tseng KY, Kuo TT, Wang V, Huang EY, Ma KH, Olson L, Hoffer BJ, Chen YH (2022) Tetrabenazine Mitigates Aberrant Release and Clearance of Dopamine in the Nigrostriatal System, and Alleviates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. Journal of Parkinson's disease 12 (5): 1545-1565. doi: https://doi.org/10.3233/jpd-223195
  8. Wang W, Jiang S, Xu C, Tang L, Liang Y, Zhao Y, Zhu G (2022) Interactions between gut microbiota and Parkinson's disease: The role of microbiota-derived amino acid metabolism. Front Aging Neurosci 14: 976316. doi: https://doi.org/10.3389/fnagi.2022.976316
  9. Choi SM, Cho SH, Kang KW, Kim JM, Kim BC (2021) Family history of hand tremor in patients with early Parkinson's disease. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia 90: 161-164. doi: https://doi.org/10.1016/j.jocn.2021.05.041
  10. Choudhury GR, Daadi MM (2018) Charting the onset of Parkinson-like motor and non-motor symptoms in nonhuman primate model of Parkinson's disease. PLoS One 13 (8): e0202770. doi: https://doi.org/10.1371/journal.pone.0202770
  11. Savitt J, Aouchiche R (2020) Management of Visual Dysfunction in Patients with Parkinson's Disease. Journal of Parkinson's disease 10 (s1): S49-s56. doi: https://doi.org/10.3233/jpd-202103
  12. Fereshtehnejad S-M, Romenets SR, Anang JB, Latreille V, Gagnon J-F, Postuma RB (2015) New clinical subtypes of Parkinson disease and their longitudinal progression: a prospective cohort comparison with other phenotypes. JAMA neurology 72 (8): 863-873. doi: https://doi.org/10.1001/jamaneurol.2015.0703
  13. Tofaris GK, Goedert M, Spillantini MG (2017) The transcellular propagation and intracellular trafficking of α-synuclein. Cold Spring Harbor perspectives in medicine 7 (9): a024380. doi: https://doi.org/10.1101%2Fcshperspect.a024380
  14. Grazia Spillantini M, Anthony Crowther R, Jakes R, Cairns NJ, Lantos PL, Goedert M (1998) Filamentous α-synuclein inclusions link multiple system atrophy with Parkinson's disease and dementia with Lewy bodies. Neuroscience Letters 251 (3): 205-208. doi: https://doi.org/10.1016/S0304-3940(98)00504-7
  15. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M (1998) α-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies. Proceedings of the National Academy of Sciences 95 (11): 6469-6473. doi: https://doi.org/10.1073/pnas.95.11.6469
  16. Furness JB, Callaghan BP, Rivera LR, Cho H-J (2014) The Enteric Nervous System and Gastrointestinal Innervation: Integrated Local and Central Control. In: Lyte M, Cryan JF (eds) Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease. Springer New York, New York, NY, pp 39-71. doi:https://doi.org/10.1007/978-1-4939-0897-4_3
  17. Qu Z-D, Thacker M, Castelucci P, Bagyanszki M, Epstein ML, Furness JB (2008) Immunohistochemical analysis of neuron types in the mouse small intestine. Cell and tissue research 334 (2): 147-161. doi: https://doi.org/10.1007/s00441-008-0684-7
  18. Gulbransen BD, Sharkey KA (2012) Novel functional roles for enteric glia in the gastrointestinal tract. Nature reviews Gastroenterology & hepatology 9 (11): 625-632. doi: https://doi.org/10.1038/nrgastro.2012.138
  19. Grundmann D, Loris E, Maas‐Omlor S, Huang W, Scheller A, Kirchhoff F, Schäfer KH (2019) Enteric glia: S100, GFAP, and beyond. The Anatomical Record 302 (8): 1333-1344. doi: https://doi.org/10.1002/ar.24128
  20. Bhukya S, S S, S S, J AQ, A A, Rani N, K D, A D, Nag TC, A S (2021) Morphological changes of the myenteric plexus at different gut segments of human fetuses. Journal of histotechnology 44 (3): 150-159. doi: https://doi.org/10.1080/01478885.2020.1862604
  21. Pan W, Rahman AA, Stavely R, Bhave S, Guyer R, Omer M, Picard N, Goldstein AM, Hotta R (2022) Schwann Cells in the Aganglionic Colon of Hirschsprung Disease Can Generate Neurons for Regenerative Therapy. Stem cells translational medicine 11 (12): 1232-1244. doi: https://doi.org/10.1093/stcltm/szac076
  22. Sanchini G, Vaes N, Boesmans W (2023) Mini-review: Enteric glial cell heterogeneity: Is it all about the niche? Neurosci Lett 812: 137396. doi: https://doi.org/10.1016/j.neulet.2023.137396
  23. Smith M, Chhabra S, Shukla R, Kenny S, Almond S, Edgar D, Wilm B (2023) The transition zone in Hirschsprung's bowel contains abnormal hybrid ganglia with characteristics of extrinsic nerves. J Cell Mol Med 27 (2): 287-298. doi: https://doi.org/10.1111/jcmm.17659
  24. Spencer NJ, Hu H (2020) Enteric nervous system: sensory transduction, neural circuits and gastrointestinal motility. Nature reviews Gastroenterology & hepatology 17 (6): 338-351. doi: https://doi.org/10.1038/s41575-020-0271-2
  25. Endres K, Schäfer K-H (2018) Influence of commensal microbiota on the enteric nervous system and its role in neurodegenerative diseases. Journal of innate immunity 10 (3): 172-180. doi: https://doi.org/10.1159/000488629
  26. Blanco AM, Calo J, Soengas JL (2021) The gut-brain axis in vertebrates: implications for food intake regulation. The Journal of experimental biology 224 (Pt 1). doi: https://doi.org/10.1242/jeb.231571
  27. Hill AE, Wade-Martins R, Burnet PWJ (2021) What Is Our Understanding of the Influence of Gut Microbiota on the Pathophysiology of Parkinson's Disease? Front Neurosci 15: 708587. doi: https://doi.org/10.3389/fnins.2021.708587
  28. Matsubara Y, Kiyohara H, Teratani T, Mikami Y, Kanai T (2022) Organ and brain crosstalk: The liver-brain axis in gastrointestinal, liver, and pancreatic diseases. Neuropharmacology 205: 108915. doi: https://doi.org/10.1016/j.neuropharm.2021.108915
  29. Navarro A, Boveris A (2009) Brain mitochondrial dysfunction and oxidative damage in Parkinson’s disease. Journal of Bioenergetics and Biomembranes 41: 517-521. doi: https://doi.org/10.1007/s10863-009-9250-6
  30. Browning KN, Carson KE (2021) Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 13 (3). doi: https://doi.org/10.3390/nu13030908
  31. Neuhuber WL, Berthoud HR (2022) Functional anatomy of the vagus system: How does the polyvagal theory comply? Biological psychology 174: 108425. doi: https://doi.org/10.1016/j.biopsycho.2022.108425
  32. van Weperen VYH, Vaseghi M (2023) Cardiac vagal afferent neurotransmission in health and disease: review and knowledge gaps. Front Neurosci 17: 1192188. doi: https://doi.org/10.3389/fnins.2023.1192188
  33. Imai J, Katagiri H (2022) Regulation of systemic metabolism by the autonomic nervous system consisting of afferent and efferent innervation. International immunology 34 (2): 67-79. doi: https://doi.org/10.1093/intimm/dxab023
  34. Minic Z, O'Leary DS, Reynolds CA (2022) Spinal Reflex Control of Arterial Blood Pressure: The Role of TRP Channels and Their Endogenous Eicosanoid Modulators. Front Physiol 13: 838175. doi: https://doi.org/10.3389/fphys.2022.838175
  35. Wachsmuth HR, Weninger SN, Duca FA (2022) Role of the gut-brain axis in energy and glucose metabolism. Experimental & molecular medicine 54 (4): 377-392. doi: https://doi.org/10.1038/s12276-021-00677-w
  36. Goronzy JJ, Li G, Yang Z, Weyand CM (2013) The janus head of T cell aging–autoimmunity and immunodeficiency. Frontiers in immunology 4: 131. doi: https://doi.org/10.3389/fimmu.2013.00131
  37. Calabrese V, Santoro A, Monti D, Crupi R, Di Paola R, Latteri S, Cuzzocrea S, Zappia M, Giordano J, Calabrese EJ (2018) Aging and Parkinson's Disease: Inflammaging, neuroinflammation and biological remodeling as key factors in pathogenesis. Free Radical Biology and Medicine 115: 80-91. doi:
  38. Mészáros Á, Molnár K, Nógrádi B, Hernádi Z, Nyúl-Tóth Á, Wilhelm I, Krizbai IA (2020) Neurovascular inflammaging in health and disease. Cells 9 (7): 1614. doi: https://doi.org/10.3390/cells9071614
  39. Zhu M, Liu X, Ye Y, Yan X, Cheng Y, Zhao L, Chen F, Ling Z (2022) Gut microbiota: a novel therapeutic target for Parkinson’s disease. Frontiers in Immunology 13: 937555. doi: https://doi.org/10.3389/fimmu.2022.937555
  40. Braak H, Tredici KD, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E (2003) Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of Aging 24 (2): 197-211. doi: https://doi.org/10.1016/S0197-4580(02)00065-9
  41. Noyce AJ, Bestwick JP, Silveira‐Moriyama L, Hawkes CH, Giovannoni G, Lees AJ, Schrag A (2012) Meta‐analysis of early nonmotor features and risk factors for Parkinson disease. Annals of neurology 72 (6): 893-901. doi: https://doi.org/10.1002/ana.23687
  42. Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A (2021) Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Parkinson's Disease 7 (1): 27. doi: https://doi.org/10.1038/s41531-021-00156-z
  43. Houser MC, Tansey MG (2017) The gut-brain axis: is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinson's disease 3 (1): 3. doi: https://doi.org/10.1038/s41531-016-0002-0
  44. Dodiya HB, Forsyth CB, Voigt RM, Engen PA, Patel J, Shaikh M, Green SJ, Naqib A, Roy A, Kordower JH, Pahan K, Shannon KM, Keshavarzian A (2020) Chronic stress-induced gut dysfunction exacerbates Parkinson's disease phenotype and pathology in a rotenone-induced mouse model of Parkinson's disease. Neurobiol Dis 135: 104352. doi: https://doi.org/10.1016/j.nbd.2018.12.012
  45. Gerhardt S, Mohajeri MH (2018) Changes of Colonic Bacterial Composition in Parkinson's Disease and Other Neurodegenerative Diseases. Nutrients 10 (6). doi: https://doi.org/10.3390/nu10060708
  46. Li C, Cui L, Yang Y, Miao J, Zhao X, Zhang J, Cui G, Zhang Y (2019) Gut Microbiota Differs Between Parkinson's Disease Patients and Healthy Controls in Northeast China. Front Mol Neurosci 12: 171. doi: https://doi.org/10.3389/fnmol.2019.00171
  47. Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A (2021) Meta-analysis of the Parkinson's disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Parkinsons Dis 7 (1): 27. doi: https://doi.org/10.1038/s41531-021-00156-z
  48. Aho VT, Pereira PA, Voutilainen S, Paulin L, Pekkonen E, Auvinen P, Scheperjans F (2019) Gut microbiota in Parkinson's disease: temporal stability and relations to disease progression. EBioMedicine 44: 691-707. doi: https://doi.org/10.1016/j.ebiom.2019.05.064
  49. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola‐Rautio J, Pohja M (2015) Gut microbiota are related to Parkinson's disease and clinical phenotype. Movement Disorders 30 (3): 350-358. doi: https://doi.org/10.1002/mds.26069
  50. Bhattarai Y, Kashyap PC (2020) Parkinson’s disease: Are gut microbes involved? American Journal of Physiology-Gastrointestinal and Liver Physiology 319 (5): G529-G540. doi: https://doi.org/10.1152/ajpgi.00058.2020
  51. Boertien JM, Pereira PA, Aho VT, Scheperjans F (2019) Increasing comparability and utility of gut microbiome studies in Parkinson’s disease: a systematic review. Journal of Parkinson's disease 9 (s2): S297-S312. doi: https://doi.org/10.3233/JPD-191711
  52. Heinzel S, Aho VT, Suenkel U, von Thaler AK, Schulte C, Deuschle C, Paulin L, Hantunen S, Brockmann K, Eschweiler GW (2021) Gut microbiome signatures of risk and prodromal markers of Parkinson disease. Annals of neurology 90 (3): E1-E12. doi: https://doi.org/10.1002/ana.26128
  53. Houser MC, Chang J, Factor SA, Molho ES, Zabetian CP, Hill‐Burns EM, Payami H, Hertzberg VS, Tansey MG (2018) Stool immune profiles evince gastrointestinal inflammation in Parkinson's disease. Movement Disorders 33 (5): 793-804. doi: https://doi.org/10.1002/mds.27326
  54. Mulak A, Koszewicz M, Panek-Jeziorna M, Koziorowska-Gawron E, Budrewicz S (2019) Fecal calprotectin as a marker of the gut immune system activation is elevated in Parkinson’s disease. Frontiers in neuroscience 13: 992. doi: https://doi.org/10.3389/fnins.2019.00992
  55. Ahrodia T, Das S, Bakshi S, Das B (2022) Structure, functions, and diversity of the healthy human microbiome. Progress in molecular biology and translational science 191 (1): 53-82. doi: https://doi.org/10.1016/bs.pmbts.2022.07.003
  56. Shao Y, Jiang Y, Li H, Zhang F, Hu Z, Zheng S (2021) Characteristics of mouse intestinal microbiota during acute liver injury and repair following 50% partial hepatectomy. Experimental and therapeutic medicine 22 (3): 953. doi: https://doi.org/10.3892/etm.2021.10385
  57. Wang D (2023) Metagenomics Databases for Bacteria. Methods in molecular biology (Clifton, NJ) 2649: 55-67. doi: https://doi.org/10.1007/978-1-0716-3072-3_3
  58. Cirstea MS, Yu AC, Golz E, Sundvick K, Kliger D, Radisavljevic N, Foulger LH, Mackenzie M, Huan T, Finlay BB (2020) Microbiota composition and metabolism are associated with gut function in Parkinson's disease. Movement Disorders 35 (7): 1208-1217. doi: https://doi.org/10.1002/mds.28052
  59. Wallen ZD, Appah M, Dean MN, Sesler CL, Factor SA, Molho E, Zabetian CP, Standaert DG, Payami H (2020) Characterizing dysbiosis of gut microbiome in PD: evidence for overabundance of opportunistic pathogens. npj Parkinson's Disease 6 (1): 11. doi: https://doi.org/10.1038/s41531-020-0112-6
  60. Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, Shibata A, Fujisawa Y, Minato T, Okamoto A (2015) Intestinal dysbiosis and lowered serum lipopolysaccharide-binding protein in Parkinson’s disease. PloS one 10 (11): e0142164. doi: https://doi.org/10.1371/journal.pone.0142164
  61. Minato T, Maeda T, Fujisawa Y, Tsuji H, Nomoto K, Ohno K, Hirayama M (2017) Progression of Parkinson's disease is associated with gut dysbiosis: two-year follow-up study. PloS one 12 (11): e0187307. doi: https://doi.org/10.1371/journal.pone.0187307
  62. Zeissig S, Bürgel N, Günzel D, Richter J, Mankertz J, Wahnschaffe U, Kroesen AJ, Zeitz M, Fromm M, Schulzke JD (2007) Changes in expression and distribution of claudin 2, 5 and 8 lead to discontinuous tight junctions and barrier dysfunction in active Crohn’s disease. Gut 56 (1): 61-72. doi: http://dx.doi.org/10.1136/gut.2006.094375
  63. Yang D, Zhao D, Ali Shah SZ, Wu W, Lai M, Zhang X, Li J, Guan Z, Zhao H, Li W (2019) The role of the gut microbiota in the pathogenesis of Parkinson's disease. Frontiers in neurology 10: 1155. doi: https://doi.org/10.3389/fneur.2019.01155
  64. Clairembault T, Leclair-Visonneau L, Coron E, Bourreille A, Le Dily S, Vavasseur F, Heymann M-F, Neunlist M, Derkinderen P (2015) Structural alterations of the intestinal epithelial barrier in Parkinson’s disease. Acta neuropathologica communications 3: 1-9. doi: https://doi.org/10.1186/s40478-015-0196-0
  65. Schwiertz A, Spiegel J, Dillmann U, Grundmann D, Bürmann J, Faßbender K, Schäfer K-H, Unger MM (2018) Fecal markers of intestinal inflammation and intestinal permeability are elevated in Parkinson's disease. Parkinsonism & Related Disorders 50: 104-107. doi: https://doi.org/10.1016/j.parkreldis.2018.02.022
  66. Forsyth CB, Shannon KM, Kordower JH, Voigt RM, Shaikh M, Jaglin JA, Estes JD, Dodiya HB, Keshavarzian A (2011) Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease. PloS one 6 (12): e28032. doi: https://doi.org/10.1371/journal.pone.0028032
  67. Trist BG, Hare DJ, Double KL (2019) Oxidative stress in the aging substantia nigra and the etiology of Parkinson's disease. Aging cell 18 (6): e13031. doi: https://doi.org/10.1111/acel.13031
  68. Ambrosi G, Cerri S, Blandini F (2014) A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. Journal of neural transmission 121: 849-859. doi: https://doi.org/10.1007/s00702-013-1149-z
  69. Lindahl M, Chalazonitis A, Palm E, Pakarinen E, Danilova T, Pham TD, Setlik W, Rao M, Võikar V, Huotari J, Kopra J, Andressoo J-O, Piepponen PT, Airavaara M, Panhelainen A, Gershon MD, Saarma M (2020) Cerebral dopamine neurotrophic factor–deficiency leads to degeneration of enteric neurons and altered brain dopamine neuronal function in mice. Neurobiology of Disease 134: 104696. doi: https://doi.org/10.1016/j.nbd.2019.104696
  70. Lee H-C, Wei Y-H (2005) Mitochondrial biogenesis and mitochondrial DNA maintenance of mammalian cells under oxidative stress. The International Journal of Biochemistry & Cell Biology 37 (4): 822-834. doi: https://doi.org/10.1016/j.biocel.2004.09.010
  71. Barodia SK, Creed RB, Goldberg MS (2017) Parkin and PINK1 functions in oxidative stress and neurodegeneration. Brain research bulletin 133: 51-59. doi: https://doi.org/10.1016/j.brainresbull.2016.12.004