Biomarker of Neuroinflammation in Parkinson’s Disease
Abstract
:1. Introduction
2. Role of Neuroinflammation in PD
3. Candidate Biomarkers of Inflammation in PDs
3.1. IL-1β
3.2. IL-2
3.3. IL-6
3.4. IL-10
3.5. TNF-α/sTNFRs
3.6. RANTES
3.7. High-Sensitivity C-Reactive Protein (hsCRP)
4. Genetic Mutations Involved in Neuroinflammation in PD
4.1. Leucine-Rich Repeat Kinase 2
4.2. PTEN-Induced Putative Kinase 1
4.3. Parkin (PRKN)
4.4. DJ-1
5. Radiotracers Targeting Microglial Activation
6. Anti-Inflammation Strategies for PDs
6.1. NSAIDs
6.2. TNF-α Inhibitor
6.3. NLRP3 Inhibitor
6.4. NRF2 Enhancer
6.5. PPAR-γ Agonist
6.6. Steroid Drugs
7. Conclusions
Candidate Biomarker | Origin | Change | Correlated Parameters | Reference |
---|---|---|---|---|
IL-1β | Serum | ↑PD | UPDRS-III, MMSE | [11] |
Serum | ↑PD with IB | [63] | ||
Serum | ↑PD | [64] | ||
Serum | ≈PD | [66] | ||
CSF | ↑PD with PRBD | [62] | ||
CSF | ≈PD | [65] | ||
IL-2 | Serum | ↑PD | MMSE | [11] |
Serum | ↑PD | [73,74] | ||
sIL-2-R | Serum | ↑PD | [75] | |
IL-6 | Serum | ↑PD | UPDRS-III | [11,78] |
Serum | ↑PD | [73,75,79,80,82,83] | ||
Serum | ↑PD with IB | [63] | ||
Serum | ↑PD | CGS, TUG | [84] | |
Serum | ↑PD with depression | [85] | ||
Serum | ↑PD mortality | [86] | ||
Serum | ↓PD | [64] | ||
Serum | ≈PD | [11,74,76,81] | ||
CSF | ↑PD | [78,80] | ||
IL-10 | Serum | ↑PD | [11,73,89] | |
Serum | ≈PD | [66,90] | ||
TNF-α | Serum | ↑PD, | UPDRS-III, MMSE | [11] |
Serum | ↑PD, | [66,75,82] | ||
Serum | ↑PD | [73] | ||
Serum | ↑PD, | Body sway, Reaction time | [83] | |
Serum | ≈PD with IB | [63] | ||
CSF | ↑PD, | [94] | ||
CSF | ↑PD with PRBD | [62] | ||
sTNFR1 | Serum | ↑PD | MMSE, Programming task of FAB | [97] |
RANTES | Serum | ↑PD | [82,100,101,102] | |
Serum | ↑PD | H&Y, disease duration | [81] | |
hsCRP | Serum | ↑PD | [106,107] | |
Plasma | ↑PD | [105] | ||
Serum | ≈PD | UPDRS-III, MMSE | [11] | |
Serum | ≈PD | [75,108] |
Target | Medication | Model | Effect | References |
---|---|---|---|---|
COX-inhibitor | Sodium salicylate | MPTP-treated mice | Beneficial | [151,152,153,155] |
Ibuprofen, piroxicam | Rotenone-treated rats | Beneficial | [154] | |
TNF-α inhibitor | Thalidomide | MPTP-treated mice | Beneficial | [159] |
NLRP3 inhibitor | MCC950 | 6-OHDA-treated mice | Beneficial | [163] |
α-Synuclein fibrils-treated mice | Beneficial | [163] | ||
NRF2 enhancer | Dimethyl fumarate | SNCA (p.A53T) transgenic mice | Beneficial | [164] |
MPTP-treated mice | Beneficial | [15] | ||
Mice expressing α-synuclein in ventral midbrain | Beneficial | [167,168] | ||
PPAR-γ agonist | Pioglitazone | MPTP-treated monkey | Beneficial | [171] |
MPTP-treated mice | Beneficial | [172,173] | ||
Rosiglitazone | MPTP-treated mice | Beneficial | [174] | |
Steroidal drugs | Dexamethasone | LPS-treated rat | Beneficial | [175] |
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Braak, H.; Del Tredici, K.; Rüb, U.; de Vos, R.A.; Jansen Steur, E.N.H.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
- Domingo, A.; Klein, C. Genetics of Parkinson disease. Handb. Clin. Neurol. 2018, 147, 211–227. [Google Scholar] [PubMed]
- Kim, C.Y.; Alcalay, R.N. Genetic forms of Parkinson’s disease. Semin Neurol. 2017, 37, 135–146. [Google Scholar] [CrossRef] [PubMed]
- Ascherio, A.; Schwarzschild, M.A. The epidemiology of Parkinson’s disease: Risk factors and prevention. Lancet Neurol. 2016, 15, 1257–1272. [Google Scholar] [CrossRef]
- De Virgilio, A.; Greco, A.; Fabbrini, G.; Inghilleri, M.; Rizzo, M.I.; Gallo, A.; Conte, M.; Rosato, C.; Appiani, M.C.; de Vincentiis, M. Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun. Rev. 2016, 15, 1005–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGeer, P.L.; Itagaki, S.; Boyes, B.E.; McGeer, E.G. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285–1291. [Google Scholar] [CrossRef]
- Gerhard, A.; Pavese, N.; Hotton, G.; Turkheimer, F.; Es, M.; Hammers, A.; Eggert, K.; Oertel, W.; Banati, R.B.; Brooks, D.J. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol. Dis. 2006, 21, 404–412. [Google Scholar] [CrossRef]
- Theodore, S.; Cao, S.; McLean, P.J.; Standaert, D.G. Targeted overexpression of human α-synuclein triggers microglial activation and an adaptive immune response in a mouse model of Parkinson disease. J. Neuropathol. Exp. Neurol. 2008, 67, 1149–1158. [Google Scholar] [CrossRef] [Green Version]
- Hong, Z.; Shi, M.; Chung, K.A.; Quinn, J.F.; Peskind, E.R.; Galasko, D.; Jankovic, J.; Zabetian, C.P.; Leverenz, J.B.; Baird, G.; et al. DJ-1 and alpha-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain 2010, 133, 713–726. [Google Scholar] [CrossRef] [Green Version]
- Parnetti, L.; Castrioto, A.; Chiasserini, D.; Persichetti, E.; Tambasco, N.; El-Agnaf, O.; Calabresi, P. Cerebrospinal fluid biomarkers in Parkinson disease. Nat. Rev. Neurol. 2013, 9, 131–140. [Google Scholar] [CrossRef]
- Williams-Gray, C.H.; Wijeyekoon, R.; Yarnall, A.J.; Lawson, R.A.; Breen, D.P.; Evans, J.R.; Cummins, G.A.; Duncan, G.W.; Khoo, T.K.; Burn, D.J.; et al. ICICLE-PD study group. Serum immune markers and disease progression in an incident Parkinson’s disease cohort (ICICLE-PD). Mov. Disord. 2016, 31, 995–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagatsu, T.; Mogi, M.; Ichinose, H.; Togari, A. Changes in cytokines and neurotrophins in Parkinson’s disease. J. Neural. Transm. Suppl. 2000, 60, 277–290. [Google Scholar]
- Hofmann, K.W.; Schuh, A.F.; Saute, J.; Townsend, R.; Fricke, D.; Leke, R.; Souza, D.O.; Portela, L.V.; Chaves, M.L.; Rieder, C.R. Interleukin-6 serum levels in patients with Parkinson’s disease. Neurochem. Res. 2009, 34, 1401–1404. [Google Scholar] [CrossRef] [Green Version]
- Scalzo, P.; Kummer, A.; Cardoso, F.; Teixeira, A.L. Increased serum levels of soluble tumor necrosis factor-alpha receptor-1 in patients with Parkinson’s disease. J. Neuroimmunol. 2009, 216, 122–125. [Google Scholar] [CrossRef] [PubMed]
- Qin, X.Y.; Zhang, S.P.; Cao, C.; Loh, Y.P.; Cheng, Y. Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: A systematic review and meta-analysis. JAMA Neurol. 2016, 73, 1316–1324. [Google Scholar] [CrossRef]
- Ezcurra, A.L.d.L.; Chertoff, M.; Ferrari, C.; Graciarena, M.; Pitossi, F. Chronic expression of low levels of tumor necrosis factor-alpha in the substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and microglia/macrophage activation. Neurobiol. Dis. 2010, 37, 630–640. [Google Scholar] [CrossRef] [PubMed]
- Imamura, K.; Hishikawa, N.; Sawada, M.; Nagatsu, T.; Yoshida, M.; Hashizume, Y. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson’s disease brains. Acta Neuropathol. 2003, 106, 518–526. [Google Scholar] [CrossRef]
- Loane, C.; Politis, M. Positron emission tomography neuroimaging in Parkinson’s disease. Am. J. Transl. Res. 2011, 3, 323–341. [Google Scholar]
- Papadopoulos, V.; Baraldi, M.; Guilarte, T.R.; Knudsen, T.B.; Lacapere, J.J.; Lindemann, P.; Norenberg, M.D.; Nutt, D.; Weizman, A.; Zhang, M.R.; et al. Translocator protein (18kDa): New nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol. Sci. 2006, 27, 402–409. [Google Scholar] [CrossRef]
- Bennett, M.C. The role of alpha-synuclein in neurodegenerative diseases. Pharmacol. Ther. 2005, 105, 311–331. [Google Scholar] [CrossRef]
- Béraud, D.; Maguire-Zeiss, K.A. Misfolded α-synuclein and toll-like receptors: Therapeutic targets for Parkinson’s disease. Parkinsonism Relat. Disord. 2012, 18, S17–S20. [Google Scholar] [CrossRef] [Green Version]
- Reynolds, A.D.; Glanzer, J.G.; Kadiu, I.; Ricardo-Dukelow, M.; Chaudhuri, A.; Ciborowski, P.; Cerny, R.; Gelman, B.; Thomas, M.P.; Mosley, R.L.; et al. Nitrated alpha-synuclein-activated microglial profiling for Parkinson’s disease. J. Neurochem. 2008, 104, 1504–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, E.J.; Woo, M.S.; Moon, P.G.; Baek, M.C.; Choi, I.Y.; Kim, W.K.; Junn, E.; Kim, H.S. Alpha-synuclein activates microglia by inducing the expressions of matrix metalloproteinases and the subsequent activation of protease-activated receptor-1. J. Immunol. 2010, 185, 615–623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Couch, Y.; Alvarez-Erviti, L.; Sibson, N.R.; Wood, M.J.; Anthony, D.C. The acute inflammatory response to intranigral α-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. J. Neuroinflammation 2011, 8, 166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panicker, N.; Sarkar, S.; Harischandra, D.S.; Neal, M.; Kam, T.I.; Jin, H.; Saminathan, H.; Langley, M.; Charli, A.; Samidurai, M.; et al. Fyn kinase regulates misfolded α-synuclein uptake and NLRP3 inflammasome activation in microglia. J. Exp. Med. 2019, 216, 1411–1430. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.; Ho, D.H.; Suk, J.E.; You, S.; Michael, S.; Kang, J.; Joong Lee, S.; Masliah, E.; Hwang, D.; Lee, H.J.; et al. Neuron-released oligomeric α-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat. Commun. 2013, 4, 1562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, I.; Zhang, Y.; Seegobin, S.P.; Pruvost, M.; Wang, Q.; Purtell, K.; Zhang, B.; Yue, Z. Microglia clear neuron-released alpha-synuclein via selective autophagy and prevent neurodegeneration. Nat. Commun. 2020, 11, 1386. [Google Scholar] [CrossRef] [Green Version]
- Choi, I.; Seegobin, S.P.; Liang, D.; Yue, Z. Synucleinphagy: A microglial “community cleanup program” for neuroprotection. Autophagy 2020, 16, 1718–1720. [Google Scholar] [CrossRef] [PubMed]
- Fellner, L.; Irschick, R.; Schanda, K.; Reindl, M.; Klimaschewski, L.; Poewe, W.; Wenning, G.K.; Stefanova, N. Toll-like receptor 4 is required for alpha-synuclein dependent activation of microglia and astroglia. Glia 2013, 61, 349–360. [Google Scholar] [CrossRef] [Green Version]
- Cebrián, C.; Zucca, F.A.; Mauri, P.; Steinbeck, J.A.; Studer, L.; Scherzer, C.R.; Kanter, E.; Budhu, S.; Mandelbaum, J.; Vonsattel, J.P.; et al. MHC-I expression renders catecholaminergic neurons susceptible to T-cell-mediated degeneration. Nat. Commun. 2014, 5, 3633. [Google Scholar] [CrossRef]
- Sanchez-Guajardo, V.; Febbraro, F.; Kirik, D.; Romero-Ramos, M. Microglia acquire distinct activation profiles depending on the degree of alpha-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS ONE 2010, 5, e8784. [Google Scholar] [CrossRef] [PubMed]
- Reynolds, A.D.; Stone, D.K.; Hutter, J.A.; Benner, E.J.; Mosley, R.L.; Gendelman, H.E. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson’s disease. J. Immunol. 2010, 184, 2261–2271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Terryberry, J.W.; Thor, G.; Peter, J.B. Autoantibodies in neurodegenerative diseases- antigen-specific frequencies and intrathecal analysis. Neurobiol. Aging 1998, 19, 205–216. [Google Scholar] [CrossRef]
- Poletaev, A.B.; Morozov, S.G.; Gnedenko, B.B.; Zlunikin, V.M.; Korzhenevskey, D.A. Serum anti-S100b, anti-GFAP and anti-NGF autoantibodies of IgG class in healthy persons and patients with mental and neurological disorders. Autoimmunity 2000, 32, 33–38. [Google Scholar] [CrossRef]
- Gruden, M.A.; Sewell, R.D.; Yanamandra, K.; Davidova, T.V.; Kucheryanu, V.G.; Bocharov, E.V.; Bocharova, O.A.; Polyschuk, V.V.; Sherstnev, V.V.; Morozova-Roche, L.A. Immunoprotection against toxic biomarkers is retained during Parkinson’s disease progression. J. Neuroimmunol. 2011, 233, 221–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karcher, D.; Federsppiel, B.S.; Lowenthal, F.D.; Frank, F.; Lowenthal, A. Anti-neurofilament antibodies in blood of patients with neurological diseases. Acta Neuropathol. 1986, 72, 82–85. [Google Scholar] [CrossRef]
- Elizan, T.S.; Casals, J.; Yahr, M.D. Antineurofilament antibodies in postencephalitic and idiopathic Parkinson’s disease. J. Neurol. Sci. 1983, 59, 341–347. [Google Scholar] [CrossRef]
- Zappia, M.; Crescibene, L.; Bosco, D.; Arabia, G.; Nicoletti, G.; Bagala, A.; Bastone, L.; Napoli, I.D.; Caracciolo, M.; Bonavita, S.; et al. Anti-GM1 ganglioside antibodies in Parkinson’s disease. Acta Neurol. Scand. 2002, 106, 54–57. [Google Scholar] [CrossRef]
- Appel, S.H.; Smith, R.G.; Alexianu, M.; Engelhardt, J.; Mosier, D.; Colom, L.; Stefani, E. Neurodegenerative disease: Autoimmunity involving calcium channels. Ann. NY Acad Sci. 1994, 747, 183–194. [Google Scholar] [CrossRef]
- Yanamandra, K.; Gruden, M.A.; Casaite, V.; Meskys, R.; Forsgren, L.; Morozova-Roche, L.A. Alpha-synuclein reactive antibodies as diagnostic biomarkers in blood sera of Parkinson’s disease patients. PLoS ONE 2011, 6, e18513. [Google Scholar] [CrossRef]
- Papachroni, K.K.; Ninkina, N.; Papapanagiotou, A.; Hadjigeorgiou, G.M.; Xiromerisiou, G.; Papadimitriou, A.; Kalofoutis, A.; Buchman, V.L. Autoantibodies to alpha-synuclein in inherited Parkinson’s disease. J. Neurochem. 2007, 101, 749–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Double, K.L.; Rowe, D.B.; Carew-Jones, F.M.; Hayes, M.; Chan, D.K.; Blackie, J.; Corbett, A.; Joffe, R.; Fung, V.S.; Morris, J.; et al. Anti-melanin antibodies are increased in sera in Parkinson’s disease. Exp. Neurol. 2009, 217, 297–301. [Google Scholar] [CrossRef] [PubMed]
- Goedert, M.; Spillantini, M.G.; Del Tredici, K.; Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 2013, 9, 13–24. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stolzenberg, E.; Berry, D.; Yang, D.; Lee, E.Y.; Kroemer, A.; Kaufman, S.; Wong, G.C.L.; Oppenheim, J.J.; Sen, S.; Fishbein, T.; et al. A role for neuronal alpha-synuclein in gastrointestinal immunity. J. Innate Immun. 2017, 9, 456–463. [Google Scholar] [CrossRef] [PubMed]
- Houser, M.C.; Chang, J.; Factor, S.A.; Molho, E.S.; Zabetian, C.P.; Hill-Burns, E.M.; Payami, H.; Hertzberg, V.S.; Tansey, M.G. Stool immune profiles evince gastrointestinal inflammation in Parkinson’s disease. Mov. Disord. 2018, 33, 793–804. [Google Scholar] [CrossRef]
- Challis, C.; Hori, A.; Sampson, T.R.; Yoo, B.B.; Challis, R.C.; Hamilton, A.M.; Mazmanian, S.K.; Volpicelli-Daley, L.A.; Gradinaru, V. Gut-seeded alpha-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat. Neurosci. 2020, 23, 327–336. [Google Scholar] [CrossRef]
- Hasegawa, S.; Goto, S.; Tsuji, H.; Okuno, T.; Asahara, T.; Nomoto, K.; Shibata, A.; Fujisawa, Y.; Minato, T.; Okamoto, A.; et al. Intestinal dysbiosis and lowered serum lipopolysaccharide-binding protein in Parkinson’s disease. PLoS ONE 2015, 10, e0142164. [Google Scholar] [CrossRef] [Green Version]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef]
- Unger, M.M.; Spiegel, J.; Dillmann, K.U.; Grundmann, D.; Philippeit, H.; Burmann, J.; Fassbender, K.; Schwiertz, A.; Schafer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Kuai, X.Y.; Yao, X.H.; Xu, L.J.; Zhou, Y.Q.; Zhang, L.P.; Liu, Y.; Pei, S.F.; Zhou, C.L. Evaluation of fecal microbiota transplantation in Parkinson’s disease patients with constipation. Microb. Cell Fact. 2021, 20, 98. [Google Scholar] [CrossRef] [PubMed]
- Parnetti, L.; Farotti, L.; Eusebi, P.; Chiasserini, D.; De Carlo, C.; Giannandrea, D.; Salvadori, N.; Lisetti, V.; Tambasco, N.; Rossi, A.; et al. Al. Differential role of CSF alpha-synuclein species, tau, and Abeta42 in Parkinson’s Disease. Front. Aging Neurosci. 2014, 6, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, L.; Tang, H.; Nie, K.; Wang, L.; Zhao, J.; Gan, R.; Huang, J.; Zhu, R.; Feng, S.; Duan, Z.; et al. Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: A systematic review and meta-analysis. Int. J. Neurosci. 2015, 125, 645–654. [Google Scholar] [CrossRef] [PubMed]
- Hall, S.; Surova, Y.; Ohrfelt, A.; Zetterberg, H.; Lindqvist, D.; Hansson, O. CSF biomarkers and clinical progression of Parkinson disease. Neurology 2015, 84, 57–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eusebi, P.; Giannandrea, D.; Biscetti, L.; Abraha, I.; Chiasserini, D.; Orso, M.; Calabresi, P.; Parnetti, L. Diagnostic utility of cerebrospinal fluid alpha-synuclein in Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2017, 32, 1389–1400. [Google Scholar] [CrossRef]
- Mollenhauer, B.; Caspell-Garcia, C.J.; Coffey, C.S.; Taylor, P.; Shaw, L.M.; Trojanowski, J.Q.; Singleton, A.; Frasier, M.; Marek, K.; Galasko, D. Longitudinal CSF biomarkers in patients with early Parkinson disease and healthy controls. Neurology 2017, 89, 1959–1969. [Google Scholar] [CrossRef] [Green Version]
- Sako, W.; Murakami, N.; Izumi, Y.; Kaji, R. Reduced alpha-synuclein in cerebrospinal fluid in synucleinopathies: Evidence from a meta-analysis. Mov. Disord. 2014, 29, 1599–1605. [Google Scholar] [CrossRef]
- Reale, M.; Iarlori, C.; Thomas, A.; Gambi, D.; Perfetti, B.; Di Nicola, M.; Onofrj, M. Peripheral cytokines profile in Parkinson’s disease. Brain Behav. 2009, 23, 55–63. [Google Scholar] [CrossRef]
- Ferrari, C.C.; Pott Godoy, M.C.; Tarelli, R.; Chertoff, M.; Depino, A.M.; Pitossi, F.J. Progressive neurodegeneration and motor disabilities induced by chronic expression of IL-1beta in the substantia nigra. Neurobiol. Dis. 2006, 24, 183–193. [Google Scholar] [CrossRef]
- Mogi, M.; Harada, M.; Narabayashi, H.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin (IL)-lβ, IL-2, IL-4, IL-6 and transforming growth factor-a levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson’s disease. Neurosci. Lett. 1996, 211, 13–16. [Google Scholar] [CrossRef]
- Nagatsu, T.; Mogi, M.; Ichinose, H.; Togari, A. Cytokines in Parkinson’s disease. J. Neural. Transm. Suppl. 2000, 10, 143–151. [Google Scholar]
- Hu, Y.; Yu, S.Y.; LJ, Z.; Cao, C.J.; Wang, F.; Chen, Z.J.; Du, Y.; Lian, T.H.; Wang, Y.J.; Chan, P.; et al. Parkinson disease with REM sleep behavior disorder: Features, α-synuclein, and inflammation. Neurology 2015, 84, 888–894. [Google Scholar] [CrossRef] [PubMed]
- Bu, X.L.; Wang, X.; Xiang, Y.; Shen, L.L.; Wang, Q.H.; Liu, Y.H.; Jiao, S.S.; Wang, Y.R.; Cao, H.Y.; Yi, X.; et al. The association between infectious burden and Parkinson’s disease: A case-control study. Parkinsonism Relat. Disord. 2015, 21, 877–881. [Google Scholar] [CrossRef] [PubMed]
- Dursun, E.; Gezen-Ak, D.; Hanagasi, H.; Bilgic, B.; Lohmann, E.; Ertan, S.; Atasoy, I.L.; Alaylioglu, M.; Araz, O.S.; Onal, B.; et al. The interleukin 1 alpha, interleukin 1 beta, interleukin 6 and alpha-2-macroglobulin serum levels in patients with early or late onset Alzheimer’s disease, mild cognitive impairment or Parkinson’s disease. J. Neuroimmunol. 2015, 283, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Pirttila, T.; Mehta, P.D.; Frey, H.; Wisniewski, H.M. Alpha 1-antichymotrypsin and IL-1 beta are not increased in CSF or serum in Alzheimer’s disease. Neurobiol. Aging 1994, 15, 313–317. [Google Scholar] [CrossRef]
- Koziorowski, D.; Tomasiuk, R.; Szlufik, S.; Friedman, A. Inflammatory cytokines and NT-proCNP in Parkinson’s disease patients. Cytokine 2012, 60, 762–766. [Google Scholar] [CrossRef]
- Lin, C.H.; Chen, C.C.; Chiang, H.L.; Liou, J.M.; Chang, C.M.; Lu, T.P.; Chuang, E.Y.; Tai, Y.C.; Cheng, C.; Lin, H.Y.; et al. Altered gut microbiota and inflammatory cytokine responses in patients with Parkinson’s disease. J. Neuroinflammation 2019, 16, 129. [Google Scholar] [CrossRef]
- Malek, T.R. The biology of interleukin-2. Annu Rev. Immunol. 2008, 26, 453–479. [Google Scholar] [CrossRef]
- Boyman, O.; Sprent, J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat. Rev. Immunol. 2012, 12, 180–190. [Google Scholar] [CrossRef]
- Sadlack, B.; Merz, H.; Schorle, H.; Schimpl, A.; Feller, A.C.; Horak, I. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 1993, 75, 253–261. [Google Scholar] [CrossRef]
- Rochman, Y.; Spolski, R.; Leonard, W.J. New insights into the regulation of t cells by gamma(c) family cytokines. Nat. Rev. Immunol. 2009, 9, 480–490. [Google Scholar] [CrossRef] [PubMed]
- Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Nagatsu, T. Interleukin-2 but not basic fibroblast growth factor is elevated in parkinsonian brain. Short communication. J. Neural. Transm. 1996, 103, 1077–1081. [Google Scholar] [CrossRef] [PubMed]
- Brodacki, B.; Staszewski, J.; Toczylowska, B.; Kozlowska, E.; Drela, N.; Chalimoniuk, M.; Stepien, A. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci. Lett. 2008, 441, 158–162. [Google Scholar] [CrossRef] [PubMed]
- Stypuła, G.; Kunert-Radek, J.; Stepień, H.; Zylińska, K.; Pawlikowski, M. Evaluation of interleukins, ACTH, cortisol and prolactin concentrations in the blood of patients with parkinson’s disease. Neuroimmunomodulation 1996, 3, 131–134. [Google Scholar] [CrossRef]
- Lindqvist, D.; Kaufman, E.; Brundin, L.; Hall, S.; Surova, Y.; Hansson, O. Non-motor symptoms in patients with Parkinson’s disease—Correlations with inflammatory cytokines in serum. PLoS ONE 2012, 7, e47387. [Google Scholar] [CrossRef] [Green Version]
- Erta, M.; Quintana, A.; Hidalgo, J. Interleukin-6, a major cytokine in the central nervous system. Int. J. Biol. Sci. 2012, 8, 1254–1266. [Google Scholar] [CrossRef]
- Gruol, D.L.; Nelson, T.E. Physiological and pathological roles of interleukin-6 in the central nervous system. Mol. Neurobiol. 1997, 15, 307–339. [Google Scholar] [CrossRef]
- Miiller, T.; Blum-Degen, D.; Przuntek, H.; Kuhn, W. Interleukin-6 levels in cerebrospinal fluid inversely correlate to severity of Parkinson’s disease. Acta Neurol. Scand. 1998, 98, 142–144. [Google Scholar] [CrossRef]
- Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci. Lett. 1994, 180, 147–150. [Google Scholar] [CrossRef]
- Blum-Degen, D.; Müller, T.; Kuhn, W.; Gerlach, M.; Przuntek, H.; Riederer, P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci. Lett. 1995, 202, 17–20. [Google Scholar] [CrossRef]
- Tang, P.; Chong, L.; Li, X.; Liu, Y.; Liu, P.; Hou, C.; Li, R. Correlation between serum RANTES levels and the severity of Parkinson’s disease. Oxid. Med. Cell Longev. 2014, 2014, 208408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gruden, M.A.; Yanamandra, K.; Kucheryanu, V.G.; Bocharova, O.R.; Sherstnev, V.V.; Morozova-Roche, L.A.; Sewell, R.D. Correlation between protective immunity to α-synuclein aggregates, oxidative stress and inflammation. Neuroimmunomodulation 2012, 19, 334–342. [Google Scholar] [CrossRef] [PubMed]
- Dobbs, R.J.; Charlett, A.; Purkiss, A.G.; Dobbs, S.M.; Weller, C.; Peterson, D.W. Association of circulating TNF-alpha and IL-6 with ageing and parkinsonism. Acta Neurol. Scand. 1999, 100, 34–41. [Google Scholar] [CrossRef] [PubMed]
- Scalzo, P.; Kummer, A.; Cardoso, F.; Teixeira, A.L. Serum levels of interleukin-6 are elevated in patients with Parkinson’s disease and correlate with physical performance. Neurosci. Lett. 2010, 468, 56–58. [Google Scholar] [CrossRef]
- Selikhova, M.V.; Kushlinskii, N.E.; Lyubimova, N.V.; Gusev, E.I. Impaired production of plasma interleukin-6 in patients with Parkinson’s disease. Bull. Exp. Biol Med. 2002, 133, 81–83. [Google Scholar] [CrossRef]
- Dufek, M.; Rektorova, I.; Thon, V.; Lokaj, J.; Rektor, I. Interleukin-6 may contribute to mortality in Parkinson’s disease patients: A 4-Year prospective study. Parkinsons Dis. 2015, 2015, 898192. [Google Scholar] [CrossRef] [Green Version]
- Moore, K.W.; O’Garra, A.; de Waal Malefyt, R.; Vieira, P.; Mosmann, T.R. Interleukin-10. Annu Rev. Immunol. 1993, 11, 165–190. [Google Scholar] [CrossRef]
- Molina-Holgado, F.; Grencis, R.; Rothwell, N.J. Actions of exogenous and endogenous IL-10 on glial responses to bacterial LPS/cytokines. Glia 2001, 33, 97–106. [Google Scholar] [CrossRef]
- Rentzos, M.; Nikolaou, C.; Andreadou, E.; Paraskevas, G.P.; Rombos, A.; Zoga, M.; Tsoutsou, A.; Boufidou, F.; Kapaki, E.; Vassilopoulos, D. Circulating interleukin-10 and interleukin-12 in Parkinson’s disease. Acta Neurol. Scand. 2009, 119, 332–337. [Google Scholar] [CrossRef]
- Rota, E.; Bellone, G.; Rocca, P.; Bergamasco, B.; Emanuelli, G.; Ferrero, P. Increased intrathecal TGF-beta1, but not IL-12, IFN-gamma and IL-10 levels in Alzheimer’s disease patients. Neurol. Sci. 2006, 27, 33–39. [Google Scholar] [CrossRef]
- Tracey, K.J.; Cerami, A. Tumor necrosis factor: A pleiotropic cytokine and therapeutic target. Annu Rev. Med. 1994, 45, 491–503. [Google Scholar] [CrossRef] [PubMed]
- Aderka, D. The potential biological and clinical significance of the soluble tumor necrosis factor receptors. Cytokine Growth Factor Rev. 1996, 7, 231–240. [Google Scholar] [CrossRef]
- Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.S.; Knapp, D.J.; Crews, F.T. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mogi, M.; Harada, M.; Riederer, P.; Narabayashi, H.; Fujita, K.; Nagatsu, T. Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett. 1994, 165, 208–210. [Google Scholar] [CrossRef]
- Teismann, P.; Tieu, K.; Cohen, O.; Choi, D.K.; Wu, D.C.; Marks, D.; Vila, M.; Jackson-Lewis, V.; Przedborski, S. Pathogenic role of glial cells in Parkinson’s disease. Mov. Disord. 2003, 18, 121–129. [Google Scholar] [CrossRef] [PubMed]
- Boka, G.; Anglade, P.; Wallach, D.; Javoy-Agid, F.; Agid, Y.; Hirsch, E.C. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neurosci. Lett. 1994, 172, 151–154. [Google Scholar] [CrossRef]
- Rocha, N.P.; Teixeira, A.L.; Scalzo, P.L.; Barbosa, I.G.; de Sousa, M.S.; Morato, I.B.; Vieira, E.L.; Christo, P.P.; Palotas, A.; Reis, H.J. Plasma levels of soluble tumor necrosis factor receptors are associated with cognitive performance in Parkinson’s disease. Mov. Disord. 2014, 29, 527–531. [Google Scholar] [CrossRef]
- Menza, M.; Dobkin, R.D.; Marin, H.; Mark, M.H.; Gara, M.; Bienfait, K.; Dicke, A.; Kusnekov, A. The role of inflammatory cytokines in cognition and oTher. non-motor symptoms of Parkinson’s disease. Psychosomatics 2010, 51, 474–479. [Google Scholar]
- Appay, V.; Rowland-Jones, S.L. RANTES: A versatile and controversial chemokine. Trends Immunol. 2001, 22, 83–87. [Google Scholar] [CrossRef]
- Mahlknecht, P.; Stemberger, S.; Sprenger, F.; Rainer, J.; Hametner, E.; Kirchmair, R.; Grabmer, C.; Scherfler, C.; Wenning, G.K.; Seppi, K.; et al. An antibody microarray analysis of serum cytokines in neurodegenerative Parkinsonian syndromes. Proteome Sci. 2012, 10, 71. [Google Scholar] [CrossRef] [Green Version]
- Rentzos, M.; Nikolaou, C.; Andreadou, E.; Paraskevas, G.P.; Rombos, A.; Zoga, M.; Tsoutsou, A.; Boufidou, F.; Kapaki, E.; Vassilopoulos, D. Circulating interleukin-15 and RANTES chemokine in Parkinson’s disease. Acta Neurol. Scand. 2007, 116, 374–937. [Google Scholar] [CrossRef] [PubMed]
- Gangemi, S.; Basile, G.; Merendino, R.A.; Epifanio, A.; Di Pasquale, G.; Ferlazzo, B.; Nicita-Mauro, V.; Morgante, L. Effect of levodopa on interleukin-15 and RANTES circulating levels in patients affected by Parkinson’s disease. Mediat. Inflamm. 2003, 12, 251–253. [Google Scholar] [CrossRef] [PubMed]
- Palasik, W.; Fiszer, U.; Lechowicz, W.; Czartoryska, B.; Krzesiewicz, M.; Lugowska, A. Assessment of relations between clinical outcome of ischemic stroke and activity of inflammatory processes in the acute phase based on examination of selected parameters. Eur. Neurol. 2005, 53, 188–193. [Google Scholar] [CrossRef] [PubMed]
- Song, I.U.; Chung, S.W.; Kim, J.S.; Lee, K.S. Association between high-sensitivity C-reactive protein and risk of early idiopathic Parkinson’s disease. Neurol. Sci. 2011, 32, 31–34. [Google Scholar] [CrossRef]
- Andican, G.; Konukoglu, D.; Bozluolcay, M.; Bayulkem, K.; Firtiina, S.; Burcak, G. Plasma oxidative and inflammatory markers in patients with idiopathic Parkinson’s disease. Acta Neurol. Belg. 2012, 112, 155–159. [Google Scholar] [CrossRef] [PubMed]
- Akil, E.; Bulut, A.; Kaplan, I.; Ozdemir, H.H.; Arslan, D.; Aluclu, M.U. The increase of carcinoembryonic antigen (CEA), high-sensitivity C-reactive protein, and neutrophil/lymphocyte ratio in Parkinson’s disease. Neurol. Sci. 2015, 36, 423–428. [Google Scholar] [CrossRef]
- Song, I.U.; Kim, Y.D.; Cho, H.J.; Chung, S.W. Is neuroinflammation involved in the development of dementia in patients with Parkinson’s disease? Intern. Med. 2013, 52, 1787–1792. [Google Scholar] [CrossRef] [Green Version]
- Song, I.U.; Kim, J.S.; Chung, S.W.; Lee, K.S. 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. Eur Neurol. 2009, 62, 99–104. [Google Scholar] [CrossRef]
- Trinh, J.; Farrer, M. Advances in the genetics of Parkinson disease. Nat. Rev. Neurol. 2013, 9, 445–454. [Google Scholar] [CrossRef]
- Paisan-Ruiz, C.; Jain, S.; Evans, E.W.; Gilks, W.P.; Simon, J.; van der Brug, M.; de Munain, A.L.; Aparicio, S.; Gil, A.M.; Khan, N.; et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 2004, 44, 595–600. [Google Scholar] [CrossRef] [Green Version]
- Zimprich, A.; Biskup, S.; Leitner, P.; Lichtner, P.; Farrer, M.; Lincoln, S.; Kachergus, J.; Hulihan, M.; Uitti, R.J.; Calne, D.B.; et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004, 44, 601–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, F.R.; Huang, W.; Chen, S.M.; Sun, L.D.; Liu, H.; Li, Y.; Cui, Y.; Yan, X.X.; Yang, H.T.; Yang, R.D.; et al. Genomewide association study of leprosy. N. Engl. J. Med. 2009, 361, 2609–2618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Umeno, J.; Asano, K.; Matsushita, T.; Matsumoto, T.; Kiyohara, Y.; Iida, M.; Nakamura, Y.; Kamatani, N.; Kubo, M. Meta-analysis of published studies identified eight additional common susceptibility loci for Crohn’s disease and ulcerative colitis. Inflamm. Bowel Dis. 2011, 17, 2407–2415. [Google Scholar] [CrossRef] [PubMed]
- Moehle, M.S.; Webber, P.J.; Tse, T.; Sukar, N.; Standaert, D.G.; DeSilva, T.M.; Cowell, R.M.; West, A.B. LRRK2 inhibition attenuates microglial inflammatory responses. J. Neurosci. 2012, 32, 1602–1611. [Google Scholar] [CrossRef] [PubMed]
- Gillardon, F.; Schmid, R.; Draheim, H. Parkinson’s disease-linked leucine-rich repeat kinase 2 (R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity. Neuroscience 2012, 208, 41–48. [Google Scholar] [CrossRef]
- Daher, J.P.; Abdelmotilib, H.A.; Hu, X.; Volpicelli-Daley, L.A.; Moehle, M.S.; Fraser, K.B.; Needle, E.; Chen, Y.; Steyn, S.J.; Galatsis, P.; et al. Leucine-rich repeat kinase 2 (LRRK2) Pharmacol. ogical inhibition abates alpha-synuclein gene-induced neurodegeneration. J. Biol. Chem 2015, 290, 19433–19444. [Google Scholar] [CrossRef] [Green Version]
- Berwick, D.C.; Heaton, G.R.; Azeggagh, S.; Harvey, K. LRRK2 Biology from structure to dysfunction: Research progresses, but the themes remain the same. Mol. Neurodegener. 2019, 14, 49. [Google Scholar] [CrossRef]
- Lin, C.H.; Lin, H.Y.; Ho, E.P.; Ke, Y.C.; Cheng, M.F.; Shiue, C.Y.; Wu, C.H.; Liao, P.H.; Hsu, A.Y.; Chu, L.A.; et al. Mild chronic colitis triggers parkinsonism in LRRK2 mutant mice through activating TNF-alpha pathway. Mov. Disord. 2021. Online ahead of print. [Google Scholar] [CrossRef]
- Dwyer, Z.; Rudyk, C.; Situt, D.; Beauchamp, S.; Abdali, J.; Dinesh, A.; Legancher, N.; Sun, H.; Schlossmacher, M.; Hayley, S. CLINT (Canadian LRRK2 in inflammation team). Microglia depletion prior to lipopolysaccharide and paraquat treatment differentially modulates behavioral and neuronal outcomes in wild type and G2019S LRRK2 knock-in mice. Brain Behav. Immun. Health 2020, 5, 100079. [Google Scholar] [CrossRef]
- Valente, E.M.; Abou-Sleiman, P.M.; Caputo, V.; Muqit, M.M.; Harvey, K.; Gispert, S.; Ali, Z.; Del Turco, D.; Bentivoglio, A.R.; Healy, D.G.; et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 2004, 304, 1158–1160. [Google Scholar] [CrossRef] [Green Version]
- Cookson, M.R. Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and oxidative stress and mitochondrial pathways. Cold Spring Harb. Perspect Med. 2012, 2, a009415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yonova-Doing, E.; Atadzhanov, M.; Quadri, M.; Kelly, P.; Shawa, N.; Musonda, S.T.; Simons, E.J.; Breedveld, G.J.; Oostra, B.A.; Bonifati, V. Analysis of LRRK2, SNCA, Parkin, PINK1, and DJ-1 in Zambian patients with Parkinson’s disease. Parkinsonism Relat. Disord. 2012, 18, 567–571. [Google Scholar] [CrossRef] [PubMed]
- Narendra, D.P.; Jin, S.M.; Tanaka, A.; Suen, D.F.; Gautier, C.A.; Shen, J.; Cookson, M.R.; Youle, R.J. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010, 8, e1000298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ziviani, E.; Tao, R.N.; Whitworth, A.J. Drosophila parkin requires PINK1 for mitochondrial translocation and ubiquitinates mitofusin. Proc. Natl. Acad. Sci. USA 2010, 107, 5018–5023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akundi, R.S.; Huang, Z.; Eason, J.; Pandya, J.D.; Zhi, L.; Cass, W.A.; Sullivan, P.G.; Bueler, H. Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1-deficient mice. PLoS ONE 2011, 6, e16038. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Byun, J.W.; Choi, I.; Kim, B.; Jeong, H.K.; Jou, I.; Joe, E. PINK1 Deficiency Enhances Inflammatory Cytokine release from acutely prepared brain slices. Exp. Neurobiol. 2013, 22, 38–44. [Google Scholar] [CrossRef] [Green Version]
- Rongvaux, A.; Jackson, R.; Harman, C.C.; Li, T.; West, A.P.; de Zoete, M.R.; Wu, Y.; Yordy, B.; Lakhani, S.A.; Kuan, C.Y.; et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell 2014, 159, 1563–1577. [Google Scholar] [CrossRef] [Green Version]
- Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef]
- White, M.J.; McArthur, K.; Metcalf, D.; Lane, R.M.; Cambier, J.C.; Herold, M.J.; van Delft, M.F.; Bedoui, S.; Lessene, G.; Ritchie, M.E.; et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 2014, 159, 1549–1562. [Google Scholar] [CrossRef] [Green Version]
- Nakahira, K.; Haspel, J.A.; Rathinam, V.A.; Lee, S.J.; Dolinay, T.; Lam, H.C.; Englert, J.A.; Rabinovitch, M.; Cernadas, M.; Kim, H.P.; et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 2011, 12, 222–230. [Google Scholar] [CrossRef] [Green Version]
- West, A.P.; Khoury-Hanold, W.; Staron, M.; Tal, M.C.; Pineda, C.M.; Lang, S.M.; Bestwick, M.; Duguay, B.A.; Raimundo, N.; MacDuff, D.A.; et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 2015, 520, 553–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitada, T.; Asakawa, S.; Hattori, N.; Matsumine, H.; Yamamura, Y.; Minoshima, S.; Yokochi, M.; Mizuno, Y.; Shimizu, N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998, 392, 605–608. [Google Scholar] [CrossRef] [PubMed]
- Shimura, H.; Hattori, N.; Kubo, S.; Mizuno, Y.; Asakawa, S.; Minoshima, S.; Shimizu, N.; Iwai, K.; Chiba, T.; Tanaka, K.; et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat. Genet. 2000, 25, 302–305. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Gao, J.; Chung, K.K.; Huang, H.; Dawson, V.L.; Dawson, T.M. Parkin functions as an E2-dependent ubiquitin- protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc. Natl Acad Sci. USA 2000, 97, 13354–13359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellosta, S.; Paoletti, R.; Corsini, A. Safety of statins: Focus on clinical pharmacokinetics and drug interactions. Circulation 2004, 109, III50–III57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feany, M.B.; Pallanck, L.J. Parkin: A multipurpose neuroprotective agent? Neuron 2003, 38, 13–16. [Google Scholar] [CrossRef] [Green Version]
- Frank-Cannon, T.C.; Tran, T.; Ruhn, K.A.; Martinez, T.N.; Hong, J.; Marvin, M.; Hartley, M.; Trevino, I.; O’Brien, D.E.; Casey, B.; et al. Parkin deficiency increases vulnerability to inflammation-related nigral degeneration. J. Neurosci. 2008, 28, 10825–10834. [Google Scholar] [CrossRef]
- Tran, T.A.; Nguyen, A.D.; Chang, J.; Goldberg, M.S.; Lee, J.K.; Tansey, M.G. Lipopolysaccharide and tumor necrosis factor regulate Parkin expression via nuclear factor-kappa B. PLoS ONE 2011, 6, e23660. [Google Scholar] [CrossRef]
- Bonifati, V.; Rizzu, P.; van Baren, M.J.; Schaap, O.; Breedveld, G.J.; Krieger, E.; Dekker, M.C.; Squitieri, F.; Ibanez, P.; Joosse, M.; et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003, 299, 256–259. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.S.; Kim, J.S.; Park, J.Y.; Suh, Y.H.; Jou, I.; Joe, E.H.; Park, S.M. DJ-1 associates with lipid rafts by palmitoylation and regulates lipid rafts-dependent endocytosis in astrocytes. Hum. Mol. Genet. 2013, 22, 4805–4817. [Google Scholar] [CrossRef] [Green Version]
- Ashley, A.K.; Hinds, A.I.; Hanneman, W.H.; Tjalkens, R.B.; Legare, M.E. DJ-1 mutation decreases astroglial release of inflammatory mediators. Neurotoxicology 2016, 52, 198–203. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.; Chen, C.; Yang, D.; Ding, J.; Wang, G.; Ren, H. DJ-1 inhibits microglial activation and protects dopaminergic neurons in vitro and in vivo through interacting with microglial p65. Cell Death Dis. 2021, 12, 715. [Google Scholar] [CrossRef] [PubMed]
- Trudler, D.; Weinreb, O.; Mandel, S.A.; Youdim, M.B.; Frenkel, D. DJ-1 deficiency triggers microglia sensitivity to dopamine toward a pro-inflammatory phenotype that is attenuated by rasagiline. J. Neurochem. 2014, 129, 434–447. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Glial reactions in Parkinson’s disease. Mov. Disord. 2008, 23, 474–483. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; He, D.; Bai, Y. Microglia-mediated inflammation and neurodegenerative disease. Mol. Neurobiol. 2016, 53, 6709–6715. [Google Scholar] [CrossRef] [PubMed]
- Banati, R.B. Visualising microglial activation in vivo. Glia 2002, 40, 206–217. [Google Scholar] [CrossRef]
- Ouchi, Y.; Yoshikawa, E.; Sekine, Y.; Futatsubashi, M.; Kanno, T.; Ogusu, T.; Torizuka, T. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann. Neurol. 2005, 57, 168–175. [Google Scholar] [CrossRef]
- Koshimori, Y.; Ko, J.H.; Mizrahi, R.; Rusjan, P.; Mabrouk, R.; Jacobs, M.F.; Christopher, L.; Hamani, C.; Lang, A.E.; Wilson, A.A.; et al. Imaging striatal microglial activation in patients with Parkinson’s disease. PLoS ONE 2015, 10, e0138721. [Google Scholar] [CrossRef]
- Hatano, K.; Yamada, T.; Toyama, H.; Kudo, G.; Nomura, M.; Suzuki, H.; Ichise, M.; Wilson, A.A.; Sawada, M.; Kato, T.; et al. Correlation between FEPPA uptake and microglia activation in 6-OHDA injured rat brain. NeuroImage 2010, 52, s138. [Google Scholar] [CrossRef]
- Ajmone-Cat, M.A.; Bernardo, A.; Greco, A.; Minghetti, L. Non-steroidal anti-Inflammatory drugs and brain inflammation: Effects on microglial functions. Pharmaceuticals 2010, 3, 1949–1965. [Google Scholar] [CrossRef]
- Sairam, K.; Saravanan, K.S.; Banerjee, R.; Mohanakumar, K.P. Non-steroidal anti-inflammatory drug sodium salicylate, but not diclofenac or celecoxib, protects against 1-methyl-4-phenyl pyridinium-induced dopaminergic neurotoxicity in rats. Brain Res. 2003, 966, 245–252. [Google Scholar] [CrossRef]
- Kurkowska-Jastrzębska, I.; Babiuch, M.; Joniec, I.; Przybyłkowski, A.; Członkowski, A.; Członkowska, A. Indomethacin protects against neurodegeneration caused by MPTP intoxication in mice. Int. Immunopharmacol. 2002, 2, 1213–1218. [Google Scholar] [CrossRef]
- Maharaj, D.S.; Saravanan, K.S.; Maharaj, H.; Mohanakumar, K.P.; Daya, S. Acetaminophen and aspirin inhibit superoxide anion generation and lipid peroxidation, and protect against 1-methyl-4-phenyl pyridinium-induced dopaminergic neurotoxicity in rats. Neurochem. Int. 2004, 44, 355–360. [Google Scholar] [CrossRef]
- Teema, A.M.; Zaitone, S.A.; Moustafa, Y.M. Ibuprofen or piroxicam protects nigral neurons and delays the development of l-dopa induced dyskinesia in rats with experimental Parkinsonism: Influence on angiogenesis. Neuropharmacology 2016, 107, 432–450. [Google Scholar] [CrossRef] [PubMed]
- Casper, D.; Yaparpalvi, U.; Rempel, N.; Werner, P. Ibuprofen protects dopaminergic neurons against glutamate toxicity in vitro. Neurosci. Lett. 2000, 289, 201–204. [Google Scholar] [CrossRef]
- Manthripragada, A.D.; Schernhammer, E.S.; Qiu, J.; Friis, S.; Wermuth, L.; Olsen, J.H.; Ritz, B. Non-steroidal anti-inflammatory drug use and the risk of Parkinson’s disease. Neuroepidemiology 2011, 36, 155–161. [Google Scholar] [CrossRef] [Green Version]
- Rees, K.; Stowe, R.; Patel, S.; Ives, N.; Breen, K.; Clarke, C.E.; Ben-Shlomo, Y. Non-steroidal anti-inflammatory drugs as disease-modifying agents for Parkinson’s disease: Evidence from observational stud. Cochrane Database Syst. Rev. 2011, 11, CD008454. [Google Scholar] [CrossRef]
- Poly, T.N.; Islam, M.M.R.; Yang, H.C.; Li, Y.J. Non-steroidal anti-inflammatory drugs and risk of Parkinson’s disease in the elderly population: A meta-analysis. Eur. J. Clin. Pharmacol. 2019, 75, 99–108. [Google Scholar] [CrossRef]
- Ferger, B.; Leng, A.; Mura, A.; Hengerer, B.; Feldon, J. Genetic ablation of tumor necrosis factor-alpha (TNF-alpha) and Pharmacol. ogical inhibition of TNF-synthesis attenuates MPTP toxicity in mouse striatum. J. Neurochem. 2004, 89, 822–833. [Google Scholar] [CrossRef]
- Peter, I.; Dubinsky, M.; Bressman, S.; Park, A.; Lu, C.; Chen, N.; Wang, A. Anti-tumor necrosis factor therapy and incidence of Parkinson disease among patients with inflammatory bowel disease. JAMA Neurol. 2018, 75, 939–946. [Google Scholar] [CrossRef]
- Pardridge, W.M. Biologic TNFα-inhibitors that cross the human blood-brain barrier. Bioeng. Bugs 2010, 1, 231–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Xia, Y.; Yin, S.; Wan, F.; Hu, J.; Kou, L.; Sun, Y.; Wu, J.; Zhou, Q.; Huang, J.; et al. Targeting microglial α-synuclein/TLRs/NF-kappaB/NLRP3 inflammasome axis in Parkinson’s disease. Front. Immunol. 2021, 12, 719807. [Google Scholar] [CrossRef] [PubMed]
- Gordon, R.; Albornoz, E.A.; Christie, D.C.; Langley, M.R.; Kumar, V.; Mantovani, S.; Robertson, A.A.B.; Butler, M.S.; Rowe, D.B.; O’Neill, L.A.; et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci. Transl. Med. 2018, 10, eaah4066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esteras, N.; Dinkova-Kostova, A.T.; Abramov, A.Y. Nrf2 activation in the treatment of neurodegenerative diseases: A focus on its role in mitochondrial bioenergetics and function. Biol Chem 2016, 397, 383–400. [Google Scholar] [CrossRef]
- Hassanzadeh, K.; Rahimmi, A. Oxidative stress and neuroinflammation in the story of Parkinson’s disease: Could targeting these pathways write a good ending? J. Cell Physiol. 2018, 234, 23–32. [Google Scholar] [CrossRef] [Green Version]
- Brandes, M.S.; Zweig, J.A.; Tang, A.; Gray, N.E. NRF2 activation ameliorates oxidative stress and improves mitochondrial function and synaptic plasticity, and in A53T α-synuclein hippocampal neurons. Antioxidants 2021, 11, 26. [Google Scholar] [CrossRef]
- Lastres-Becker, I.; Garcia-Yague, A.J.; Scannevin, R.H.; Casarejos, M.J.; Kugler, S.; Rabano, A.; Cuadrado, A. Repurposing the NRF2 activator dimethyl fumarate as therapy against synucleinopathy in Parkinson’s disease. Antioxid. Redox Signal. 2016, 25, 61–77. [Google Scholar] [CrossRef] [Green Version]
- Campolo, M.; Casili, G.; Biundo, F.; Crupi, R.; Cordaro, M.; Cuzzocrea, S.; Esposito, E. The neuroprotective effect of dimethyl fumarate in an MPTP-mouse model of Parkinson’s disease: Involvement of reactive oxygen species/nuclear factor-kappaB/nuclear transcription factor related to NF-E2. Antioxid. Redox Signal. 2017, 27, 453–471. [Google Scholar] [CrossRef] [Green Version]
- Delerive, P.; Fruchart, J.C.; Staels, B. Peroxisome proliferator-activated receptors in inflammation control. J. Endocrinol 2001, 169, 453–459. [Google Scholar] [CrossRef] [Green Version]
- Chaturvedi, R.K.; Beal, M.F. PPAR: A therapeutic target in Parkinson’s disease. J. Neurochem. 2008, 106, 506–518. [Google Scholar] [CrossRef]
- Swanson, C.R.; Joers, V.; Bondarenko, V.; Brunner, K.; Simmons, H.A.; Ziegler, T.E.; Kemnitz, J.W.; Johnson, J.A.; Emborg, M.E. The PPAR-gamma agonist pioglitazone modulates inflammation and induces neuroprotection in parkinsonian monkeys. J. Neuroinflammation 2011, 8, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Breidert, T.; Callebert, J.; Heneka, M.T.; Landreth, G.; Launay, J.M.; Hirsch, E.C. Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson’s disease. J. Neurochem. 2002, 82, 615–624. [Google Scholar] [CrossRef] [PubMed]
- Dehmer, T.; Heneka, M.T.; Sastre, M.; Dichgans, J.; Schulz, J.B. Protection by pioglitazone in the MPTP model of Parkinson’s disease correlates with I kappa B alpha induction and block of NF kappa B and iNOS activation. J. Neurochem. 2004, 88, 494–501. [Google Scholar] [CrossRef] [PubMed]
- Schintu, N.; Frau, L.; Ibba, M.; Caboni, P.; Garau, A.; Carboni, E.; Carta, A.R. PPAR-gamma-mediated neuroprotection in a chronic mouse model of Parkinson’s disease. Eur. J. Neurosci. 2009, 29, 954–963. [Google Scholar] [CrossRef]
- Castano, A.; Herrera, A.J.; 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-a, IL-1b and IFN-c. J. Neurochem. 2002, 81, 150–157. [Google Scholar] [CrossRef]
- Bourque, M.; Di Paolo, T. Neuroactive steroids and Parkinson’s disease. Curr. Opin. Endocr. Metab. Res. 2022, 22, 100312. [Google Scholar] [CrossRef]
- Frau, R.; Miczán, V.; Traccis, F.; Aroni, S.; Pongor, C.I.; Saba, P.; Serra, V.; Sagheddu, C.; Fanni, S.; Congiu, M.; et al. Prenatal THC exposure produces a hyperdopaminergic phenotype rescued by pregnenolone. Nat. Neurosci. 2019, 22, 1975–1985. [Google Scholar] [CrossRef] [Green Version]
- Belanger, N.; Gregoire, L.; Bedard, P.J.; Di Paolo, T. DHEA improves symptomatic treatment of moderately and severely impaired MPTP monkeys. Neurobiol. Aging 2006, 27, 1684–1693. [Google Scholar] [CrossRef]
- Maclagan, L.C.; Visanji, N.P.; Cheng, Y.; Tadrous, M.; Lacoste, A.M.B.; Kalia, L.V.; Bronskill, S.E.; Marras, C. Identifying drugs with disease-modifying potential in Parkinson’s disease using artificial intelligence and pharmacoepidemiology. Pharmacoepidemiol. Drug Saf. 2020, 29, 864–872. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Liu, T.-W.; Chen, C.-M.; Chang, K.-H. Biomarker of Neuroinflammation in Parkinson’s Disease. Int. J. Mol. Sci. 2022, 23, 4148. https://doi.org/10.3390/ijms23084148
Liu T-W, Chen C-M, Chang K-H. Biomarker of Neuroinflammation in Parkinson’s Disease. International Journal of Molecular Sciences. 2022; 23(8):4148. https://doi.org/10.3390/ijms23084148
Chicago/Turabian StyleLiu, Tsai-Wei, Chiung-Mei Chen, and Kuo-Hsuan Chang. 2022. "Biomarker of Neuroinflammation in Parkinson’s Disease" International Journal of Molecular Sciences 23, no. 8: 4148. https://doi.org/10.3390/ijms23084148
APA StyleLiu, T. -W., Chen, C. -M., & Chang, K. -H. (2022). Biomarker of Neuroinflammation in Parkinson’s Disease. International Journal of Molecular Sciences, 23(8), 4148. https://doi.org/10.3390/ijms23084148