Evidence of inflammatory system involvement in Parkinson's disease

Yinxia Chao, Siew Cheng Wong, Eng King Tan, Yinxia Chao, Siew Cheng Wong, Eng King Tan

Abstract

Parkinson's disease (PD) is a chronic neurodegenerative disease underpinned by both genetic and environmental etiologic factors. Recent findings suggest that inflammation may be a pathogenic factor in the onset and progression of both familial and sporadic PD. Understanding the precise role of inflammatory factors in PD will likely lead to understanding of how the disease arises. In vivo evidence for inflammation in PD includes dysregulated molecular mediators such as cytokines, complement system and its receptors, resident microglial activation, peripheral immune cells invasion, and altered composition and phenotype of peripheral immune cells. The growing awareness of these factors has prompted novel approaches to modulate the immune system, although it remains whether these approaches can be used in humans. Influences of ageing and differential exposure to environmental agents suggest potential host-pathogen specific pathophysiologic factors. There is a clear need for research to further unravel the pathophysiologic role of immunity in PD, with the potential of developing new therapeutic targets for this debilitating condition.

Figures

Figure 1
Figure 1
The paper is summarized in the schematic drawing. Genetic mutations and environmental exposures can selectively induce dopaminergic neuron loss in the SNc. The mechanism may involve both regional (microglia) and systematic (peripheral) immune dysregulation. The dysregulated immune system can either elicit or exacerbate dopaminergic neuron loss at SNc by direct contact or by overrelease of cytokines and other immune mediators and thus immune regulation may have great therapeutic potential in PD treatment.

References

    1. Dauer W, Przedborski S. Parkinson's disease: mechanisms and models. Neuron. 2003;39(6):889–909.
    1. Członkowska A, Kurkowska-Jastrzebska I, Członkowski A, Peter D, Stefano GB. Immune processes in the pathogenesis of Parkinson’s disease—a potential role for microglia and nitric oxide. Medical Science Monitor. 2002;8(8):RA165–RA177.
    1. Whitton PS. Inflammation as a causative factor in the aetiology of Parkinson's disease. British Journal of Pharmacology. 2007;150(8):963–976.
    1. Kim DK, Kim HS, Kim A, et al. DJ-1 regulates mast cell activation and IgE-mediated allergic responses. Journal of Allergy and Clinical Immunology. 2013;131(6):1653–1662.
    1. Manzanillo PS, Ayres JS, Watson RO, et al. The ubiquitin ligase parkin mediates resistance to intracellular pathogens. Nature. 2013;501(7468):512–516.
    1. Deleidi M, Hallett PJ, Koprich JB, Chung C, Isacson O. The toll-like receptor-3 agonist polyinosinic:polycytidylic acid triggers nigrostriatal dopaminergic degeneration. Journal of Neuroscience. 2010;30(48):16091–16101.
    1. Qin L, Liu Y, Hong J, Crews FT. NADPH oxidase and aging drive microglial activation, oxidative stress, and dopaminergic neurodegeneration following systemic LPS administration. GLIA. 2013;61(6):855–868.
    1. Tanji K, Mori F, Imaizumi T, et al. Upregulation of α-synuclein by lipopolysaccharide and interleukin-1 in human macrophages. Pathology International. 2002;52(9):572–577.
    1. Russo I, Bubacco L, Greggio E. LRRK2 and neuroinflammation: partners in crime in Parkinson's disease? Journal of Neuroinflammation. 2014;11, article 52
    1. Brodacki B, Staszewski J, Toczyłowska B, et al. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFα, and INFγ concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neuroscience Letters. 2008;441(2):158–162.
    1. Reale M, Greig NH, Kamal MA. Peripheral chemo-cytokine profiles in Alzheimer's and Parkinson's diseases. Mini-Reviews in Medicinal Chemistry. 2009;9(10):1229–1241.
    1. Song IU, Kim JS, Chung SW, Lee KS. Is there an association between the level of high-sensitivity C-reactive protein and idiopathic parkinson's disease? A comparison of parkinson's disease patients, disease controls and healthy individuals. European Neurology. 2009;62(2):99–104.
    1. Mogi M, Harada M, Kondob T, et al. Interleukin-1β, interleukin-6, epidermal growth factor and transforming growth factor-α are elevated in the brain from parkinsonian patients. Neuroscience Letters. 1994;180(2):147–150.
    1. Mogi M, Harada M, Kondo T, Riederer P, Nagatsu T. Brain β 2-microglobulin levels are elevated in the striatum in Parkinson's diseaselevels are elevated in the striatum in Parkinson's disease. Journal of Neural Transmission: Parkinson's Disease and Dementia Section. 1995;9(1):87–92.
    1. Blum-Degen D, Müller T, Kuhn W. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neuroscience Letters. 1995;202:17–20.
    1. Mount MP, Lira A, Grimes D, et al. Involvement of interferon-γ in microglial-mediated loss of dopaminergic neurons. Journal of Neuroscience. 2007;27(12):3328–3337.
    1. Kim E, Hwang O. Role of matrix metalloproteinase-3 in neurodegeneration. Journal of Neurochemistry. 2011;116(1):22–32.
    1. Chung YC, Kim Y, Bok E, Yune TY, Maeng S, Jin BK. MMP-3 contributes to nigrostriatal dopaminergic neuronal loss, BBB damage, and neuroinflammation in an MPTP mouse model of Parkinson's disease. Mediators of Inflammation. 2013;2013:11 pages.370526
    1. Yamada S, Nagai T, Nakai T, et al. Matrix metalloproteinase-3 is a possible mediator of neurodevelopmental impairment due to polyI: C-induced innate immune activation of astrocytes. Brain, Behavior, and Immunity. 2014;38:272–282.
    1. Fagundes CT, Costa VV, Cisalpino D, et al. IFN-γ production depends on IL-12 and IL-18 combined action and mediates host resistance to dengue virus infection in a nitric oxide-dependent manner. PLoS Neglected Tropical Diseases. 2011;5(12, article e1449)
    1. Kannan Y, Yu J, Raices RM, et al. IκBζ augments IL-12- and IL-18-mediated IFN-γ production in human NK cells. Blood. 2011;117(10):2855–2863.
    1. Takahashi HK, Iwagaki H, Hamano R, et al. Effects of adenosine on adhesion molecule expression and cytokine production in human PBMC depend on the receptor subtype activated. British Journal of Pharmacology. 2007;150(6):816–822.
    1. Gillett A, Hedreul MT, Khademi M, et al. Interleukin 18 receptor 1 expression distinguishes patients with multiple sclerosis. Multiple Sclerosis. 2010;16(9):1056–1065.
    1. Millward JM, Løbner M, Wheeler RD, Owens T. Inflammation in the central nervous system and Th17 responses are inhibited by IFN-γ-induced IL-18 binding protein. The Journal of Immunology. 2010;185(4):2458–2466.
    1. Celik SK, Öz ZS, Dursun A, et al. Interleukin 18 gene polymorphism is a risk factor for multiple sclerosis. Molecular Biology Reports. 2014;41(3):1653–1658.
    1. Sugama S, Wirz SA, Barr AM, Conti B, Bartfai T, Shibasaki T. Interleukin-18 null mice show diminished microglial activation and reduced dopaminergic neuron loss following acute 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine treatment. Neuroscience. 2004;128(2):451–458.
    1. Xu X, Li D, He Q, Gao J, Chen B, Xie A. Interleukin-18 promoter polymorphisms and risk of Parkinson's disease in a Han Chinese population. Brain Research. 2011;1381:90–94.
    1. Nie K, Zhang Y, Gan R, et al. Polymorphisms in immune/inflammatory cytokine genes are related to Parkinson's disease with cognitive impairment in the Han Chinese population. Neuroscience Letters. 2013;541:111–115.
    1. Yamada T, McGeer PL, McGeer EG. Lewy bodies in Parkinson's disease are recognized by antibodies to complement proteins. Acta Neuropathologica. 1992;84(1):100–104.
    1. Koziorowski D, Tomasiuk R, Szlufik S, Friedman A. Inflammatory cytokines and NT-proCNP in Parkinson's disease patients. Cytokine. 2012;60(3):762–766.
    1. Béraud D, Hathaway HA, Trecki J, et al. Microglial activation and antioxidant responses induced by the Parkinson's disease protein α-synuclein. Journal of Neuroimmune Pharmacology. 2013;8(1):94–117.
    1. Iwamura C, Nakayama T. Toll-like receptors in the respiratory system: their roles in inflammation. Current Allergy and Asthma Reports. 2008;8(1):7–13.
    1. Cario E. Toll-like receptors in inflammatory bowel diseases: a decade later. Inflammatory Bowel Diseases. 2010;16(9):1583–1597.
    1. Seneviratne AN, Sivagurunathan B, Monaco C. Toll-like receptors and macrophage activation in atherosclerosis. Clinica Chimica Acta. 2012;413(1-2):3–14.
    1. Brudek T, Winge K, Agander TK, Pakkenberg B. Screening of toll-like receptors expression in multiple system atrophy brains. Neurochemical Research. 2013;38(6):1252–1259.
    1. Roodveldt C, Labrador-Garrido A, Gonzalez-Rey E, et al. Preconditioning of microglia by α-synuclein strongly affects the response induced by toll-like receptor (TLR) stimulation. PLoS ONE. 2013;8(11)e79160
    1. Saresella M, Marventano I, Calabrese E, et al. A complex proinflammatory role for peripheral monocytes in Alzheimer’s disease. Journal of Alzheimer’s Disease. 2014;38(2):403–413.
    1. Hafner-Bratkovič I, Benčina M, Fitzgerald KA, Golenbock D, Jerala R. NLRP3 inflammasome activation in macrophage cell lines by prion protein fibrils as the source of IL-1β and neuronal toxicity. Cellular and Molecular Life Sciences. 2012;69(24):4215–4228.
    1. Schnaars M, Beckert H, Halle A. Assessing β-amyloid-induced NLRP3 inflammasome activation in primary microglia. Methods in Molecular Biology. 2013;1040:1–8.
    1. Tan M, Yu J, Jiang T, Zhu X, Tan L. The NLRP3 inflammasome in Alzheimer's disease. Molecular Neurobiology. 2013;48(3):875–882.
    1. Meng XF, Wang XL, Tian XJ, et al. Nod-like receptor protein 1 inflammasome mediates neuron injury under high glucose. Molecular Neurobiology. 2014;49(2):673–684.
    1. Devine MJ, Kaganovich A, Ryten M, et al. Pathogenic LRRK2 mutations do not alter gene expression in cell model systems or human brain tissue. PLoS ONE. 2011;6(7)e22489
    1. Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature. 2001;414(6866):916–920.
    1. Saha N, Moldovan F, Tardif G, et al. Interleukin-1β-converting enzyme (caspase-1) in human osteoarthritic tissues. Localization and role in the maturation of interleukin-1β and interleukin-18. Arthritis & Rheumatism. 1999;42(8):1577–1587.
    1. Sullivan GW. Adenosine A2A receptor agonists as anti-inflammatory agents. Current Opinion in Investigational Drugs. 2003;4(11):1313–1319.
    1. Yamagata T, Ichinose M. Agents against cytokine synthesis or receptors. European Journal of Pharmacology. 2006;533(1–3):289–301.
    1. Vincenzi F, Corciulo C, Targa M, et al. Multiple sclerosis lymphocytes upregulate A2A adenosine receptors that are antiinflammatory when stimulated. European Journal of Immunology. 2013;43(8):2206–2216.
    1. Burnstock G, Fredholm BB, Verkhratsky A. Adenosine and ATP receptors in the brain. Current Topics in Medicinal Chemistry. 2011;11(8):973–1011.
    1. Casetta I, Vincenzi F, Bencivelli D, et al. A2A adenosine receptors and Parkinson's disease severity. Acta Neurologica Scandinavica. 2014;129(4):276–281.
    1. Khanapur S, Waarde AV, Ishiwata K, et al. Adenosine A2A receptor Antagonists as Positron Emission Tomography (PET) Tracers. Current Medicinal Chemistry. 2013;21(3):312–328.
    1. Fox SH. Non-dopaminergic treatments for motor control in Parkinson's disease. Drugs. 2013;73(13):1405–1415.
    1. Chao YX, He BP, Tay SS. Mesenchymal stem cell transplantation attenuates blood brain barrier damage and neuroinflammation and protects dopaminergic neurons against MPTP toxicity in the substantia nigra in a model of Parkinson's disease. Journal of Neuroimmunology. 2009;216(1-2):39–50.
    1. Prineas JW, Parratt JDE. Oligodendrocytes and the early multiple sclerosis lesion. Annals of Neurology. 2012;72(1):18–31.
    1. Zhou Y, Wang Y, Kovacs M, Jin J, Zhang J. Microglial activation induced by neurodegeneration: a proteomic analysis. Molecular and Cellular Proteomics. 2005;4(10):1471–1479.
    1. Smeyne M, Jiao Y, Shepherd KR, Smeyne RJ. Glia cell number modulates sensitivity to MPTP in mice. Glia. 2005;52(2):144–152.
    1. Sawada M, Imamura K, Nagatsu T. Role of cytokines in inflammatory process in Parkinson's disease. Journal of Neural Transmission, Supplement. 2006;(70):373–381.
    1. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38(8):1285–1291.
    1. Gerhard A, Pavese N, Hotton G, et al. In vivo imaging of microglial activation with 11C(R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiology of Disease. 2006;21(2):404–412.
    1. Członkowska A, Kohutnicka M, Kurkowska-Jastrzȩbska I, Członkowski A. Microglial reaction in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced Parkinson's disease mice model. Neurodegeneration. 1996;5(2):137–143.
    1. Kohutnicka M, Lewandowska E, Kurkowska-Jastrzebska I, Członkowski A, Członkowska A. Microglial and astrocytic involvement in a murine model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Immunopharmacology. 1998;39(3):167–180.
    1. Lee Mosley R, Benner EJ, Kadiu I, et al. Neuroinflammation, oxidative stress, and the pathogenesis of Parkinson's disease. Clinical Neuroscience Research. 2006;6(5):261–281.
    1. Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D. Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Annals of Neurology. 1999;46(4):598–605.
    1. Hamza TH, Zabetian CP, Tenesa A, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nature Genetics. 2010;42(9):781–785.
    1. Chock VY, Giffard RG. Development of neonatal murine microglia In vitro: changes in response to lipopolysaccharide and ischemia-like injury. Pediatric Research. 2005;57(4):475–480.
    1. Lee M. Neurotransmitters and microglial-mediated neuroinflammation. Current Protein and Peptide Science. 2013;14(1):21–32.
    1. Pais TF, Szegő ÉM, Marques O, et al. The NAD-dependent deacetylase sirtuin 2 is a suppressor of microglial activation and brain inflammation. EMBO Journal. 2013;32(19):2603–2616.
    1. Mclaughlin P, Zhou Y, Ma T, et al. Proteomic analysis of microglial contribution to mouse strain-dependent dopaminergic neurotoxicity. Glia. 2006;53(6):567–582.
    1. de Lella Ezcurra AL, Chertoff M, Ferrari C, Graciarena M, Pitossi F. Chronic expression of low levels of tumor necrosis factor-α in the substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and microglia/macrophage activation. Neurobiology of Disease. 2010;37(3):630–640.
    1. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)-1β, IL-2, IL-4, IL-6 and transforming growth factor-α levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease. Neuroscience Letters. 1996;211(1):13–16.
    1. Ferrari CC, Pott Godoy MC, Tarelli R, Chertoff M, Depino AM, Pitossi FJ. Progressive neurodegeneration and motor disabilities induced by chronic expression of IL-1β in the substantia nigra. Neurobiology of Disease. 2006;24(1):183–193.
    1. Godoy MCP, Tarelli R, Ferrari CC, Sarchi MI, Pitossi FJ. Central and systemic IL-1 exacerbates neurodegeneration and motor symptoms in a model of Parkinson's disease. Brain. 2008;131, part 7:1880–1894.
    1. Gordon R, Anantharam V, Kanthasamy AG. Proteolytic activation of proapoptotic kinase protein kinase Cδ by tumor necrosis factor α death receptor signaling in dopaminergic neurons during neuroinflammation. Journal of Neuroinflammation. 2012;9(article 82)
    1. Sriram K, Matheson JM, Benkovic SA, Miller DB, Luster MI, O'Callaghan JP. Deficiency of TNF receptors suppresses microglial activation and alters the susceptibility of brain regions to MPTP-induced neurotoxicity: role of TNF-α1. FASEB Journal. 2006;20(6):670–682.
    1. Stone DK, Reynolds AD, Mosley RL, Gendelman HE. Innate and adaptive immunity for the pathobiology of Parkinson's disease. Antioxidants and Redox Signaling. 2009;11(9):2151–2166.
    1. Appel SH, Beers DR, Henkel JS. T cell-microglial dialogue in Parkinson's disease and amyotrophic lateral sclerosis: are we listening? Trends in Immunology. 2010;31(1):7–17.
    1. Harm AS, Cao S, Rowse AL, et al. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. Journal of Neuroscience. 2013;33(23):9592–9600.
    1. Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: effects of oligomeric and fibrillar amyloid-β . Journal of Neuroimmunology. 2009;210(1-2):3–12.
    1. Engelhardt B. The blood-central nervous system barriers actively control immune cell entry into the central nervous system. Current Pharmaceutical Design. 2008;14(16):1555–1565.
    1. Alvarez JI, Dodelet-Devillers A, Kebir H, et al. The Hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence. Science. 2011;334(6063):1727–1731.
    1. McGeer PL, Kawamata T, Walker DG, Akiyama H, Tooyama I, McGeer EG. Microglia in degenerative neurological disease. Glia. 1993;7(1):84–92.
    1. Brochard V, Combadière B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. Journal of Clinical Investigation. 2009;119(1):182–192.
    1. Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson’s disease. Parkinson's Disease. 2011;2011:25 pages.487450
    1. Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson's disease. Journal of Leukocyte Biology. 2007;82(5):1083–1094.
    1. Reynolds AD, Stone DK, Hutter JAL, Benner EJ, Mosley RL, Gendelman HE. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson's disease. Journal of Immunology. 2010;184(5):2261–2271.
    1. Bartels AL, Willemsen ATM, Kortekaas R, et al. Decreased blood-brain barrier P-glycoprotein function in the progression of Parkinson's disease, PSP and MSA. Journal of Neural Transmission. 2008;115(7):1001–1009.
    1. Appay V, Rowland-Jones SL. RANTES: a versatile and controversial chemokine. Trends in Immunology. 2001;22(2):83–87.
    1. Rentzos M, Nikolaou C, Andreadou E, et al. Circulating interleukin-15 and RANTES chemokine in Parkinson's disease. Acta Neurologica Scandinavica. 2007;116(6):374–379.
    1. Stevens CH, Rowe D, Morel-Kopp M, et al. Reduced T helper and B lymphocytes in Parkinson's disease. Journal of Neuroimmunology. 2012;252(1-2):95–99.
    1. Niwa F, Kuriyama N, Nakagawa M, Imanishi J. Effects of peripheral lymphocyte subpopulations and the clinical correlation with Parkinson's disease. Geriatrics and Gerontology International. 2012;12(1):102–107.
    1. Funk N, Wieghofer P, Grimm S, et al. Characterization of peripheral hematopoietic stem cells and monocytes in Parkinson's disease. Movement Disorders. 2013;28(3):392–395.
    1. Shin EC, Cho SE, Lee D, et al. Expression patterns of α-synuclein in human hematopoietic cells and in Drosophila at different developmental stages. Molecules and Cells. 2000;10(1):65–70.
    1. Su AI, Wiltshire T, Batalov S, et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(16):6062–6067.
    1. Austin SA, Floden AM, Murphy EJ, Combs CK. α-synuclein expression modulates microglial activation phenotype. Journal of Neuroscience. 2006;26(41):10558–10563.
    1. Wu C, Orozco C, Boyer J, et al. BioGPS: an extensible and customizable portal for querying and organizing gene annotation resources. Genome Biology. 2009;10(11, article R130)
    1. Cardoso CC, Pereira AC, de Sales Marques C, Moraes MO. Leprosy susceptibility: genetic variations regulate innate and adaptive immunity, and disease outcome. Future Microbiology. 2011;6(5):533–549.
    1. Trabzuni D, Ryten M, Emmett W, et al. Fine-mapping, gene expression and splicing analysis of the disease associated LRRK2 locus. PLoS ONE. 2013;8(8)e70724
    1. Hirotani M, Maita C, Niino M, et al. Correlation between DJ-1 levels in the cerebrospinal fluid and the progression of disabilities in multiple sclerosis patients. Multiple Sclerosis. 2008;14(8):1056–1060.
    1. Semchuk KM, Love EJ, Lee RG. Parkinson’s disease: a test of the multifactorial etiologic hypothesis. Neurology. 1993;43(6):1173–1180.
    1. Vlajinac H, Dzoljic E, Maksimovic J, Marinkovic J, Sipetic S, Kostic V. Infections as a risk factor for Parkinson's disease: a case-control study. International Journal of Neuroscience. 2013;123(5):329–332.
    1. Tomé CML, Tyson T, Rey NL, Grathwohl S, Britschgi M, Brundin P. Inflammation and α-synuclein's prion-like behavior in Parkinson's disease—is there a link? Molecular Neurobiology. 2013;47(2):561–574.
    1. Olanow CW, Brundin P. Parkinson's disease and alpha synuclein: is Parkinson's disease a prion-like disorder? Movement Disorders. 2013;28(1):31–40.
    1. Lee CS, Won C, Yoo H, et al. Inhibition of double-stranded RNA-induced inducible nitric oxide synthase expression by fraxinellone and sauchinone in murine microglia. Biological and Pharmaceutical Bulletin. 2009;32(11):1870–1874.
    1. Ha D, Stone DK, Mosley RL, Gendelman HE. Immunization strategies for Parkinson's disease. Parkinsonism and Related Disorders. 2012;18(1):S218–S221.
    1. McCoy MK, Ruhn KA, Martinez TN, McAlpine FE, Blesch A, Tansey MG. Intranigral lentiviral delivery of dominant-negative TNF attenuates neurodegeneration and behavioral deficits in hemiparkinsonian rats. Molecular Therapy. 2008;16(9):1572–1579.
    1. Harms AS, Barnum CJ, Ruhn KA, et al. Delayed dominant-negative TNF gene therapy halts progressive loss of nigral dopaminergic neurons in a rat model of parkinson's disease. Molecular Therapy. 2011;19(1):46–52.
    1. Chen H, Zhang SM, Hernán MA, et al. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Archives of Neurology. 2003;60(8):1059–1064.
    1. Etminan M, Carleton BC, Samii A. Non-steroidal anti-inflammatory drug use and the risk of Parkinson disease: a retrospective cohort study. Journal of Clinical Neuroscience. 2008;15(5):576–577.
    1. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of parkinsons disease: systematic review and meta-analysis of observational studies. Drugs & Aging. 2009;26(9):769–779.
    1. Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen and risk of Parkinson disease. Neurology. 2011;76(10):863–869.
    1. Liu B, Du L, Hong JS. Naloxone protects rat dopaminergic neurons against inflammatory damage through inhibition of microglia activation and superoxide generation. The Journal of Pharmacology and Experimental Therapeutics. 2000;293(2):607–617.
    1. Castaño A, Herrera AJ, Cano J, Machado A. The degenerative effect of a single intranigral injection of LPS on the dopaminergic system is prevented by dexamethasone, and not mimicked by rh-TNF-α IL-1β IFN-γ . Journal of Neurochemistry. 2002;81(1):150–157.
    1. Kim H, Suh Y. Minocycline and neurodegenerative diseases. Behavioural Brain Research. 2009;196(2):168–179.
    1. Lu L, Li F, Wang X. Novel anti-inflammatory and neuroprotective agents for Parkinson's disease. CNS and Neurological Disorders—Drug Targets. 2010;9(2):232–240.
    1. Lee M, Tazzari V, Giustarini D, et al. Effects of hydrogen sulfide-releasing L-DOPA derivatives on glial activation: potential for treating Parkinson disease. The Journal of Biological Chemistry. 2010;285(23):17318–17328.
    1. Sánchez-Pernaute R, Ferree A, Cooper O, Yu M, Brownell A, Isacson O. Selective COX-2 inhibition prevents progressive dopamine neuron degeneration in a rat model of Parkinson's disease. Journal of Neuroinflammation. 2004;1(1, article 6)
    1. Teismann P. COX-2 in the neurodegenerative process of Parkinson's disease. BioFactors. 2012;38(6):395–397.
    1. Paul G, Anisimov SV. The secretome of mesenchymal stem cells: potential implications for neuroregeneration. Biochimie. 2013;95(12):2246–2256.

Source: PubMed

3
구독하다