The Universal Non-Neuronal Nature of Parkinson's Disease: A Theory

Main Article Content

Andre X.C.N. Valente
Altynai Adilbayeva
Tursonjan Tokay
Albert Rizvanov

Abstract

Parkinson's disease (PD) is one of the most common neurodegenerative disorders, yet the etiology of the majority of its cases remains unknown. In this manuscript, relevant published evidence is interpreted and integrated into a comprehensive hypothesis on the nature, origin, and inter-cellular mode of propagation of sporadic PD. We propose to characterize sporadic PD as a pathological deviation in the global gene expression program of a cell: the PD expression-state, or PD-state for short. A universal cell-generic state, the PD-state deviation would be particularly damaging in a neuronal context, ultimately leading to neuron death and the ensuing observed clinical signs. We review why ageing associated accumulated damage caused by oxidative stress in mitochondria could be the trigger for a primordial cell to shift to the PD-state. We propose that hematopoietic cells could be the first to acquire the PD-state, at hematopoiesis, from the disruption in reactive oxygen species homeostasis that arises with age in the hematopoietic stem-cell niche. We argue that cellular ageing is nevertheless unlikely to explain the shift to the PD-state of all the subsequently affected cells in a patient, thus indicating the existence of a distinct mechanism of cellular propagation of the PD-state. We highlight recently published findings on the inter-cellular exchange of mitochondrial DNA and the ability of mitochondrial DNA to modulate the cellular global gene expression state and propose this could form the basis for the inter-cellular transmission of the PD-state.

Article Details

How to Cite
Valente, A. X., Adilbayeva, A., Tokay, T., & Rizvanov, A. (2016). The Universal Non-Neuronal Nature of Parkinson’s Disease: A Theory. Central Asian Journal of Global Health, 5(1). https://doi.org/10.5195/cajgh.2016.231
Section
Perspective

References

Statistics on Parkinson's.

Shrestha LB. Population aging in developing countries. Health Aff. Vol 19 (3)2000:204-212.

Davie CA. A review of Parkinson's disease. Vol 862008:109-127.

Beitz JM. Parkinson's disease: A review. Front Biosci. Vol 62014:65-74.

Imputation of sequence variants for identification of genetic risks for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet. Vol 377 (9766)2011:641–649.

Nalls MA, Pankratz N, Lill CM, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson's disease. Nat Genet. Vol 462014:989–993.

Monte DAD, Lavasani M, Manning-Bog AB. Environmental factors in Parkinson's disease. Neurotoxicology. Vol 23 (4-5)2002:487-502.

Takahashi M, Yamada T. Viral etiology for Parkinson's disease - a possible role of influenza A virus infection. Jpn J Infect Dis. Vol 52 (3)1999:89-98.

Olanow CW, Prusiner SB. Is Parkinson's disease a prion disorder? Proc Natl Acad Sci U S A. Vol 106 (31)2009:12571-12572.

Lerner A, Bagic A. Olfactory pathogenesis of idiopathic Parkinson disease revisited. Mov Disord. Vol 23 (8)2008:1076-1084.

Phillips RJ, Walter GC, Wilder SL, Baronowsky EA, Powley TL. Alpha-synuclein-immunopositive myenteric neurons and vagal preganglionic terminals: autonomic pathway implicated in Parkinson's disease? Neuroscience. Vol 153 (3)2008:733-750.

Hawkes CH, Del Tredici K, Braak H. Parkinson's disease: a dual-hit hypothesis. Neuropath Appl Neuro. Vol 33 (6)2007:599-614.

Desplats P, Lee H-J, Bae E-J, Patrick C, Rockenstein E, Crews L. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of α-synuclein. Vol 106 (31)2009:13010-13015.

Henchcliffe C, Beal MF. Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat Clin Pract Neuro. Vol 4 (11)2008:600-609.

Swerdlow RH. Does Mitochondrial DNA Play a Role in Parkinson's Disease? A Review of Cybrid and Other Supportive Evidence. Antioxid Redox Signal. Vol 16(9)2012:950-964.

Monahan AJ, Warren M, Carvey PM. Neuroinflammation and Peripheral Immune Infiltration in Parkinson's Disease: An Autoimmune Hypothesis. Cell Transplant. Vol 17 (4)2008:363-372.

Whitton P. Inflammation as a causative factor in the aetiology of Parkinson's disease. Br J Pharmacol. Vol 150 (8)2007:963-976.

Valente AXCN, Sousa JAB, Outeiro TF, Ferreira L. A stem-cell ageing hypothesis on the origin of Parkinson's disease. In: Valente AXCN, ed. Science and engineering in high-throughput biology including a theory on Parkinson's disease: Lulu Books; 2011:43-73.

Sánchez-Danés A, Richaud-Patin Y, Carballo-Carbajal I, et al. Disease-specific phenotypes in dopamine neurons from human iPS-based models of genetic and sporadic Parkinson's Disease. EMBO Mol Med. Vol 42012:380-395.

Woodard CM, Campos BA, Kuo S-H, et al. iPSC-Derived dopamine neurons reveal differences between monozygotic twins discordant for Parkinson's disease. Cell Reports. Vol 92014:1173-1182.

Tanner CM, Ottman R, Goldman SM, et al. Parkinson disease in twins: an etiologic study. JAMA - J Am Med Assoc. Vol 2811999:341-346.

Wiredefeldt K, Gatz M, Reynolds CA, Prescott CA, Pedersen NL. Heritability of Parkinson disease in Swedish twins: a longitudinal study. Neurobiol Aging. Vol 322011:1923.e1921-1923.e1928.

Valente AXCN, Oliveira PJ, Khaiboullina SF, Palotás A, Rizvanov AA. Biological Insight, High-Throughput Datasets and the Nature of Neuro-Degenerative Diseases. Curr Drug Metab. Vol 14 (7)2013:814-818.

Simunovic F, Yi M, Wang Y, et al. Gene expression profiling of substantia nigra dopamine neurons: further insights into Parkinson's disease pathology. Brain. Vol 1322009:1795-1809.

Scherzer CR, Eklund AC, Morse LJ, et al. Molecular markers of early Parkinson's disease based on gene expression in blood. Proc Natl Acad Sci U S A. Vol 104 (3)2007:955–960.

Mandel S, Grunblatt E, Riederer P, et al. Gene Expression Profiling of Sporadic Parkinson's Disease Substantia Nigra Pars Compacta Reveals Impairment of Ubiquitin-Proteasome Subunits, SKP1A, Aldehyde Dehydrogenase, and Chaperone HSC-70. Ann N Y Acad Sci. Vol 10532008:356–375.

Masliah E, W. Dumaop DG, Desplats P. Distinctive patterns of DNA methylation associated with Parkinson disease. Epigenetics. Vol 8 (10)2013:1030-1038.

Lane N. A unifying view of ageing and disease: the double-agent theory. J Theor Biol. Vol 225 (4)2003:531-540.

Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. Vol 4172009:1-13.

Harmann D. Aging - a theory based on free-radical and radiation chemistry. J Gerontol. Vol 111956:298-300.

Barja G. Updating the mitochondrial free-radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. Vol 192013:1420-1445.

Wiedemann FR, Winkler K, Lins H, Wallesch C-W, Kunz WS. Detection of respiratory chain defects in cultivated skin fibroblasts and skeletal muscle of patients with Parkinson's disease. Ann N Y Acad Sci. Vol 8932006:426-429.

Barroso N, Campos Y, Huertas R, Esteban J, Molina JA. Respiratory chain enzyme activities in lymphocytes from untreated patients with Parkinson disease. Clin Chem. Vol 391993:667-669.

Yoshino H, Nakagawa-Hattori Y, Kondo T, Mizuno Y. Mitochondrial complex I and II activities of lymphocytes and platelets in Parkinson's disease. J Neural Transm Vol 4 (1)1992:27-34.

Mytilineou C, Werner P, Molinari S, Rocco AD, Cohen G, Yahr MD. Impaired oxidative decarboxylation of pyruvate in fibroblasts from patients with Parkinson's disease. J Neural Transm. Vol 8 (3)1994:223-228.

Braak H, Tredici KD, Rüb U, Vos RAId, Steur ENHJ, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology Aging. Vol 242003:197–211.

Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. Vol 142008:501-503.

Kordower JH, Chun Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med. Vol 142008:504-506.

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. Vol 92007:654 - 659.

Tan AS, Baty JW, Dong L-F, et al. Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA. Cell Metab. Vol 21 (1)2015:81-94.

Jayaprakash AD, Benson EK, Gone S, et al. Stable heteroplasmy at the single-cell level is facilitated by intercellular exchange of mtDNA. Nucleic Acids Res. Vol 43 (4)2015:2177-2187.

Trimmer PA, Borland MK, Keeney PM, Jr. JPB, Jr. WDP. Parkinson's disease transgenic mitochondrial cybrids generate Lewy inclusion bodies. Vol 882004:800-812.

Esteves AR, Domingues AF, Ferreira IL, et al. Mitochondrial function in Parkinson's disease cybrids containing an nt2 neuron-like nuclear background. Mitochondrion. Vol 82008:219-228.

Smiraglia DJ, Kulawiec M, Bistulfi GL, Gupta SG, Singh KK. A novel role for mitochondria in regulating epigenetic modification in the nucleus. Cener Biol Ther. Vol 72008:1182-1190.

Xie CH, Naito A, Mizumachi T, Evans TT, Douglas MG, Cooney CA. Mitochondrial regulation of cancer associated nuclear DNA methylation. Biochem Biophys Res Commun. Vol 656-6612007:364.

Bellizzi D, D'Aquila P, Giordano M, Montesanto A, Passarino G. Global DNA methylation levels are modulated by mitochondrial DNA variants. Epigenomics. Vol 42012:17-27.

Kelly RD, Rodda AE, Dickinson A, et al. Mitochondrial DNA haplotypes define gene expression patterns in pluripotent and differentiating emrbyonic stem cells. Stem Cells. Vol 312013:703-716.

Schapira AHV. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol. Vol 72008:97-109.

Maresca A, Zaffagnini M, Caporali L, Carelli V, Zanna C. DNA methyltransferase 1 mutations and mitochondrial pathology: is mtDNA methylated? Front Genet. Vol 6 art. no. 92015.

Ghosh S, Singh KK, Sengupta S, Scaria V. Mitoepigenetics: the different shades of grey. Mitochondrion. Vol 252015:60-66.

Hong EE, Okitsu CY, Smith AD, Hsieh C-L. Regionally specific and genome-wide analyses conclusively demonstrate the absence of CpG methylation in humann mitochondrial DNA. Mol Cell Biol. Vol 33 (14)2013:2683-2690.

Bacarelli AA, Byun H-M. Platelet mitochondrial DNA methylation: a potential new marker of cardiovascular disease. Clinical Epigenetics. Vol 7 (44)2015.

Stuart MJ, Murphy S, Oski FA. A simple nonradioisotope technic for the determination of platelet life-span. N Engl J Med. Vol 2921975:1310-1313.

Simon SI, Kim M-H. A day (or 5) in a neutrophil's life. Blood. Vol 116 (4)2010:511 - 512.

Tough DF, Sprent J. Lifespan of lymphocytes. Immunol Res. Vol 14 (1)1995:1-12.

Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. Vol 5 (8)2007:e201.

Rossi DJ, Bryder D, Zahn JM, et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci USA. Vol 1022005:9194-9199.

Hamanaka RB, Chandel NS. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci. Vol 35 (9)2010:505–513.

Pervaiz S, Taneja R, Ghaffari S. Oxidative stress regulation of stem and progenitor cells. Antioxid Redox Signal. Vol 11(11)2009:2777-2789.

Francis R, Richardson C. Multipotent hematopoietic cells susceptible to alternative double-strand break repair pathways that promote genome rearrangements. Genes Dev. Vol 212007:1064-1074.

Shao L, Luo Y, Zhou D. Hematopoietic stem-cell injury induced by ionizing radiation. Antioxid Redox Signal. Vol 202014:1447-1462.

Owusu-Ansah E, Banerjee U. Reactive oxygen species prime drosophila hematopoietic progenitors for differentiation. Nature. Vol 4612009:537-541.

Yahata T, Takanashi T, Muguruma Y, et al. Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood. Vol 118 (11)2011:2941-2950.

Chen J, Astle CM, Harrison DE. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp Hematol. Vol 312003:1097-1103.

Ito K, Hirao A, Arai F, et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. Vol 122006:446–451.

Brown K, Xie S, Qiu X, et al. SIRT3 reverses aging-associated degeneration. Cell Rep. Vol 32013:319-327.

Tothova Z, Kollipara R, Huntly BJ, et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. Vol 1282007:325–339.

Abbas HA, Maccio DR, Coskun S, Jackson JG, Hazen AL, Sills TM. Mdm2 is required for survival of hematopoietic stem cells/progenitors via dampening of ROS-induced p53 activity. Cell Stem Cell. Vol 72010:606-617.

Yalcin S, Marinkovic D, Mungamuri SK, et al. ROS‐mediated amplification of AKT/mTOR signalling pathway leads to myeloproliferative syndrome in Foxo3−/− mice. EMBO J. Vol 29 (24)2010:4118–4131.

Ito K, Hirao A, Arai F, et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem-cells. Nature. Vol 4312004:997-1002.

Xiao W, Shameli A, Harding CV, Meyerson HJ, Maitta RW. Late stages of hematopoiesis and B cell lymphopoiesis are regulated by α-synuclein, a key player in Parkinson's Disease. Immunobiology. Vol 2192014:836-844.

Valente AXCN, Shin JH, Sarkar A, Gao Y. Rare coding SNP in DZIP1 gene associated with late-onset sporadic Parkinson's disease. Sci Rep. Vol 2 art. no. 2562012.

Sekimizu K, Nishioka N, Sasaki H, Takeda H, Karlstrom RO, Kawakami A. The zebrafish iguana locus encodes Dzip1, a novel zinc-finger protein required for proper regulation of Hedgehog signaling. Vol 131 (11)2004:2521-2532.

Palma V, Lim DA, Dahmane N, Sánchez P, Brionne TC. Sonic hedgehog controls stem cell behavior in the postnatal and adult brain. Development. Vol 132 (2)2004:335-344.

Bhardwaj G, Murdoch B, Wu D, Baker DP, Williams KP. Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nature Immunol. Vol 22001:172-180.

Sidransky E, Nalls MA, Aasly JO, et al. Multicenter Analysis of Glucocerebrosidase Mutations in Parkinson's Disease. N Engl J Med. Vol 3612009:1651-1661.

Mingyi C, Jun W. Gaucher Disease: Review of the Literature. Arch Pathol Lab Med. Vol 132.52008:851-853.

Dawson TM. New Animal Models for Parkinson's Disease. Cell. Vol 101 (2)2000:115–118.

Natale G, Pasquali L, Ruggieri SAP, Fornai F. Parkinson's disease and the gut: a well known clinical association in need of an effective cure and explanation. Neurogastroenterol Motil. Vol 20 (7)2008:741-749.

Creamer B, Shorter RG, Bamforth J. The turnover and shedding of epithelial cells. I. The turnover in the gastro-intestinal tract. Gut. Vol 2 (2)1961:110-116.

Mouret A, Lepousez G, Gras J, Gabellec MM, Lledo PM. Turnover of newborn olfactory bulb neurons optimizes olfaction. J Neurosci. Vol 29 (39)2009:12302-12314.