Alpha-Synuclein and Microglia in Parkinson’s Disease: From Pathogenesis to Therapeutic Prospects
Abstract
:1. Introduction
2. Role of α-Syn in PD
2.1. α-Syn: Structure and Physiological Function
2.2. Modification of α-Syn in PD: Triggers of Aggregation and Disease Progression
3. Interplay Between α-Syn and Microglia in PD
3.1. Recognition of α-Syn Aggregates by Microglia
3.2. Propagation of α-Syn Aggregates
3.3. Activation of Inflammatory Pathways by α-Syn Aggregates
3.4. Clearance of α-Syn Aggregates
3.4.1. Clearance of α-Syn Aggregates Through Phagocytosis and Autophagy in Microglia
3.4.2. Impairment of α-Syn Clearance Pathways in PD
4. Therapeutic Approaches and Future Directions
4.1. Targeting α-Syn Propagation Pathways
4.2. Modulating Microglial Activation States
Drugs | Therapeutic Strategy | Efficacy | Status | clinicaltrials.gov ID |
---|---|---|---|---|
Neuroinflammation | ||||
Minocycline | Inhibition of microglial activation | Reduced inflammation in α-syn mouse model [117]; no significant therapeutic effect in early PD [118,119]. | Phase II | NCT00063193 (completed) |
NPT520-34 | Inhibition of microglial activation | Confirmed safety, tolerability, and pharmacokinetic profile in healthy volunteers; planned to evaluate efficacy in PD. | Phase I | NCT03954600 (completed) |
TAK-242 | TLR4 inhibitor | Shown to suppress microglial activation and reduce neuroinflammation in preclinical models of PD by targeting TLR4 [112]. | Preclinical | |
CU-CPT22 | TLR2 inhibitor | Reduced p-α-syn-induced inflammation and restored autophagic function in an MPTP mouse model [84,114]. | Preclinical | |
Inzomelid | NLRP3 inflammasome inhibitor | Entered Phase I clinical trials as a potential disease-modifying therapy for PD. | Phase I | NCT04015076 (completed) |
α-syn clearance | ||||
Ambroxol | Enhances GCase activity | Increased GCase levels and reduced α-syn accumulation in both patients with and without GBA1 mutations [122]. | Phase II | NCT02941822 (completed) NCT06193421 (recruiting) NCT02914366 (Active, not recruiting) |
BIA 28-6156 | Allosteric activator of GCase | Undergoing evaluation to determine efficacy in patients with GBA1 gene variant. | Phase II | NCT05819359 (Active, not recruiting) |
Venglustat (GZ/SAR402671) | Glucosylceramide synthase inhibitor | Reduced α-syn in preclinical models but failed to show efficacy in Phase II trials, leading to discontinuation. | Phase II | NCT02906020 (Terminated) |
PR001 (LY3884961) | Enhances GCase activity | Undergoing evaluation to assess the safety, tolerability, immune response, biomarkers, and efficacy in patients. | Phase I/II | NCT04127578 (Recruiting) |
ATC161 | Enhances autophagy | Reduced brain α-syn aggregates and glial inflammation, improving motor function in a PD model [124]. | Preclinical | |
α-syn propagation | ||||
GW4869 | Neutral sphingomyelinase inhibitor (blocks exosome release) | Reduced α-syn propagation in preclinical studies by inhibiting the release of exosome-contained α-synuclein [111]. | Preclinical | - |
4.3. Personalized and Gene Therapy Approaches
4.4. Diagnosing and Monitoring PD Progression Using PET Imaging
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Global, regional, and national burden of neurological disorders during 1990-2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2017, 16, 877–897. [CrossRef] [PubMed]
- Zhu, J.; Cui, Y.; Zhang, J.; Yan, R.; Su, D.; Zhao, D.; Wang, A.; Feng, T. Temporal trends in the prevalence of Parkinson’s disease from 1980 to 2023: A systematic review and meta-analysis. Lancet Healthy Longev. 2024, 5, e464–e479. [Google Scholar] [CrossRef] [PubMed]
- Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Bendor, J.T.; Logan, T.P.; Edwards, R.H. The function of α-synuclein. Neuron 2013, 79, 1044–1066. [Google Scholar] [CrossRef]
- Maroteaux, L.; Campanelli, J.T.; Scheller, R.H. Synuclein: A neuron-specific protein localized to the nucleus and presynaptic nerve terminal. J. Neurosci. 1988, 8, 2804–2815. [Google Scholar] [CrossRef]
- Burré, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Südhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef]
- Zaltieri, M.; Grigoletto, J.; Longhena, F.; Navarria, L.; Favero, G.; Castrezzati, S.; Colivicchi, M.A.; Della Corte, L.; Rezzani, R.; Pizzi, M.; et al. α-synuclein and synapsin III cooperatively regulate synaptic function in dopamine neurons. J. Cell Sci. 2015, 128, 2231–2243. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.H.C.; Wislet-Gendebien, S.; Samuel, F.; Visanji, N.P.; Zhang, G.; Marsilio, D.; Langman, T.; Fraser, P.E.; Tandon, A. α-Synuclein membrane association is regulated by the Rab3a recycling machinery and presynaptic activity. J. Biol. Chem. 2013, 288, 7438–7449. [Google Scholar] [CrossRef]
- Brás, I.C.; Outeiro, T.F. Alpha-Synuclein: Mechanisms of Release and Pathology Progression in Synucleinopathies. Cells 2021, 10, 375. [Google Scholar] [CrossRef]
- Tofaris, G.K. Initiation and progression of α-synuclein pathology in Parkinson’s disease. Cell. Mol. Life Sci. 2022, 79, 210. [Google Scholar] [CrossRef]
- Zhang, W.; Wang, T.; Pei, Z.; Miller, D.S.; Wu, X.; Block, M.L.; Wilson, B.; Zhang, W.; Zhou, Y.; Hong, J.S.; et al. Aggregated alpha-synuclein activates microglia: A process leading to disease progression in Parkinson’s disease. FASEB J. 2005, 19, 533–542. [Google Scholar] [CrossRef]
- Zhang, Q.S.; Heng, Y.; Yuan, Y.H.; Chen, N.H. Pathological α-synuclein exacerbates the progression of Parkinson’s disease through microglial activation. Toxicol. Lett. 2017, 265, 30–37. [Google Scholar] [CrossRef] [PubMed]
- Thi Lai, T.; Kim, Y.E.; Nguyen, L.T.N.; Thi Nguyen, T.; Kwak, I.H.; Richter, F.; Kim, Y.J.; Ma, H.I. Microglial inhibition alleviates alpha-synuclein propagation and neurodegeneration in Parkinson’s disease mouse model. NPJ Park. Dis. 2024, 10, 32. [Google Scholar] [CrossRef]
- Lv, Q.K.; Tao, K.X.; Wang, X.B.; Yao, X.Y.; Pang, M.Z.; Liu, J.Y.; Wang, F.; Liu, C.F. Role of α-synuclein in microglia: Autophagy and phagocytosis balance neuroinflammation in Parkinson’s disease. Inflamm. Res. 2023, 72, 443–462. [Google Scholar] [CrossRef] [PubMed]
- Badanjak, K.; Fixemer, S.; Smajić, S.; Skupin, A.; Grünewald, A. The Contribution of Microglia to Neuroinflammation in Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 4676. [Google Scholar] [CrossRef]
- Zheng, T.; Zhang, Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson’s disease. Neurochem. Int. 2021, 148, 105094. [Google Scholar] [CrossRef]
- Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate α-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef]
- Guo, M.; Hao, Y.; Feng, Y.; Li, H.; Mao, Y.; Dong, Q.; Cui, M. Microglial Exosomes in Neurodegenerative Disease. Front. Mol. Neurosci. 2021, 14, 630808. [Google Scholar] [CrossRef]
- Abounit, S.; Bousset, L.; Loria, F.; Zhu, S.; de Chaumont, F.; Pieri, L.; Olivo-Marin, J.C.; Melki, R.; Zurzolo, C. Tunneling nanotubes spread fibrillar α-synuclein by intercellular trafficking of lysosomes. EMBO J. 2016, 35, 2120–2138. [Google Scholar] [CrossRef]
- McFarthing, K.; Buff, S.; Rafaloff, G.; Pitzer, K.; Fiske, B.; Navangul, A.; Beissert, K.; Pilcicka, A.; Fuest, R.; Wyse, R.K.; et al. Parkinson’s Disease Drug Therapies in the Clinical Trial Pipeline: 2024 Update. J. Park. Dis. 2024, 14, 899–912. [Google Scholar] [CrossRef] [PubMed]
- Menon, S.; Armstrong, S.; Hamzeh, A.; Visanji, N.P.; Sardi, S.P.; Tandon, A. Alpha-Synuclein Targeting Therapeutics for Parkinson’s Disease and Related Synucleinopathies. Front. Neurol. 2022, 13, 852003. [Google Scholar] [CrossRef] [PubMed]
- Rodger, A.T.; ALNasser, M.; Carter, W.G. Are Therapies That Target α-Synuclein Effective at Halting Parkinson’s Disease Progression? A Systematic Review. Int. J. Mol. Sci. 2023, 24, 11022. [Google Scholar] [CrossRef] [PubMed]
- Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
- Dixit, S.; Bohre, K.; Singh, Y.; Himeur, Y.; Mansoor, W.; Atalla, S.; Srinivasan, K. A Comprehensive Review on AI-Enabled Models for Parkinson’s Disease Diagnosis. Electronics 2023, 12, 783. [Google Scholar] [CrossRef]
- Xiang, J.; Tao, Y.; Xia, Y.; Luo, S.; Zhao, Q.; Li, B.; Zhang, X.; Sun, Y.; Xia, W.; Zhang, M.; et al. Development of an α-synuclein positron emission tomography tracer for imaging synucleinopathies. Cell 2023, 186, 3350–3367.e3319. [Google Scholar] [CrossRef]
- Gupta, R.; Kumari, S.; Senapati, A.; Ambasta, R.K.; Kumar, P. New era of artificial intelligence and machine learning-based detection, diagnosis, and therapeutics in Parkinson’s disease. Ageing Res. Rev. 2023, 90, 102013. [Google Scholar] [CrossRef]
- Bhandari, N.; Walambe, R.; Kotecha, K.; Kaliya, M. Integrative gene expression analysis for the diagnosis of Parkinson’s disease using machine learning and explainable AI. Comput. Biol. Med. 2023, 163, 107140. [Google Scholar] [CrossRef]
- Emamzadeh, F.N. Alpha-synuclein structure, functions, and interactions. J. Res. Med. Sci. 2016, 21, 29. [Google Scholar] [CrossRef]
- Del Mar, C.; Greenbaum, E.A.; Mayne, L.; Englander, S.W.; Woods, V.L., Jr. Structure and properties of alpha-synuclein and other amyloids determined at the amino acid level. Proc. Natl. Acad. Sci. USA 2005, 102, 15477–15482. [Google Scholar] [CrossRef]
- Rajagopalan, S.; Andersen, J.K. Alpha synuclein aggregation: Is it the toxic gain of function responsible for neurodegeneration in Parkinson’s disease? Mech. Ageing Dev. 2001, 122, 1499–1510. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, J.A.; Ivanova, M.I.; Sawaya, M.R.; Cascio, D.; Reyes, F.E.; Shi, D.; Sangwan, S.; Guenther, E.L.; Johnson, L.M.; Zhang, M.; et al. Structure of the toxic core of α-synuclein from invisible crystals. Nature 2015, 525, 486–490. [Google Scholar] [CrossRef] [PubMed]
- Lavedan, C. The synuclein family. Genome Res. 1998, 8, 871–880. [Google Scholar] [CrossRef] [PubMed]
- Sode, K.; Ochiai, S.; Kobayashi, N.; Usuzaka, E. Effect of reparation of repeat sequences in the human alpha-synuclein on fibrillation ability. Int. J. Biol. Sci. 2006, 3, 1–7. [Google Scholar] [CrossRef]
- Samuel, F.; Flavin, W.P.; Iqbal, S.; Pacelli, C.; Sri Renganathan, S.D.; Trudeau, L.E.; Campbell, E.M.; Fraser, P.E.; Tandon, A. Effects of Serine 129 Phosphorylation on α-Synuclein Aggregation, Membrane Association, and Internalization. J. Biol. Chem. 2016, 291, 4374–4385. [Google Scholar] [CrossRef]
- Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
- Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef]
- Lesage, S.; Anheim, M.; Letournel, F.; Bousset, L.; Honoré, A.; Rozas, N.; Pieri, L.; Madiona, K.; Dürr, A.; Melki, R.; et al. G51D α-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann. Neurol. 2013, 73, 459–471. [Google Scholar] [CrossRef]
- Krüger, R.; Kuhn, W.; Müller, T.; Woitalla, D.; Graeber, M.; Kösel, S.; Przuntek, H.; Epplen, J.T.; Schöls, L.; Riess, O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 106–108. [Google Scholar] [CrossRef]
- Zarranz, J.J.; Alegre, J.; Gómez-Esteban, J.C.; Lezcano, E.; Ros, R.; Ampuero, I.; Vidal, L.; Hoenicka, J.; Rodriguez, O.; Atarés, B.; et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004, 55, 164–173. [Google Scholar] [CrossRef]
- Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J.; et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 2013, 28, 811–813. [Google Scholar] [CrossRef] [PubMed]
- Proukakis, C.; Dudzik, C.G.; Brier, T.; MacKay, D.S.; Cooper, J.M.; Millhauser, G.L.; Houlden, H.; Schapira, A.H. A novel α-synuclein missense mutation in Parkinson disease. Neurology 2013, 80, 1062–1064. [Google Scholar] [CrossRef] [PubMed]
- Pasanen, P.; Myllykangas, L.; Siitonen, M.; Raunio, A.; Kaakkola, S.; Lyytinen, J.; Tienari, P.J.; Pöyhönen, M.; Paetau, A. Novel α-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol. Aging 2014, 35, 2180.e1–2180.e5. [Google Scholar] [CrossRef] [PubMed]
- Hassanzadeh, K.; Liu, J.; Maddila, S.; Mouradian, M.M. Post-translational Modifications of α-Synuclein, their Therapeutic Potential and Crosstalk in Health and Neurodegenerative Diseases. Pharmacol. Rev. 2024, 76, 1254–1290. [Google Scholar] [CrossRef] [PubMed]
- Okochi, M.; Walter, J.; Koyama, A.; Nakajo, S.; Baba, M.; Iwatsubo, T.; Meijer, L.; Kahle, P.J.; Haass, C. Constitutive phosphorylation of the Parkinson’s disease associated alpha-synuclein. J. Biol. Chem. 2000, 275, 390–397. [Google Scholar] [CrossRef]
- Fujiwara, H.; Hasegawa, M.; Dohmae, N.; Kawashima, A.; Masliah, E.; Goldberg, M.S.; Shen, J.; Takio, K.; Iwatsubo, T. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat. Cell Biol. 2002, 4, 160–164. [Google Scholar] [CrossRef]
- Danielson, S.R.; Held, J.M.; Schilling, B.; Oo, M.; Gibson, B.W.; Andersen, J.K. Preferentially increased nitration of alpha-synuclein at tyrosine-39 in a cellular oxidative model of Parkinson’s disease. Anal. Chem. 2009, 81, 7823–7828. [Google Scholar] [CrossRef]
- Benner, E.J.; Banerjee, R.; Reynolds, A.D.; Sherman, S.; Pisarev, V.M.; Tsiperson, V.; Nemachek, C.; Ciborowski, P.; Przedborski, S.; Mosley, R.L.; et al. Nitrated alpha-synuclein immunity accelerates degeneration of nigral dopaminergic neurons. PLoS ONE 2008, 3, e1376. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Qiang, M.; Wei, Y.; He, R. A novel molecular mechanism for nitrated {alpha}-synuclein-induced cell death. J. Mol. Cell. Biol. 2011, 3, 239–249. [Google Scholar] [CrossRef] [PubMed]
- Trexler, A.J.; Rhoades, E. N-Terminal acetylation is critical for forming α-helical oligomer of α-synuclein. Protein Sci. 2012, 21, 601–605. [Google Scholar] [CrossRef]
- Bell, R.; Thrush, R.J.; Castellana-Cruz, M.; Oeller, M.; Staats, R.; Nene, A.; Flagmeier, P.; Xu, C.K.; Satapathy, S.; Galvagnion, C.; et al. N-Terminal Acetylation of α-Synuclein Slows down Its Aggregation Process and Alters the Morphology of the Resulting Aggregates. Biochemistry 2022, 61, 1743–1756. [Google Scholar] [CrossRef] [PubMed]
- Shimura, H.; Schlossmacher, M.G.; Hattori, N.; Frosch, M.P.; Trockenbacher, A.; Schneider, R.; Mizuno, Y.; Kosik, K.S.; Selkoe, D.J. Ubiquitination of a new form of alpha-synuclein by parkin from human brain: Implications for Parkinson’s disease. Science 2001, 293, 263–269. [Google Scholar] [CrossRef] [PubMed]
- Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [PubMed]
- Sierra, A.; Beccari, S.; Diaz-Aparicio, I.; Encinas, J.M.; Comeau, S.; Tremblay, M. Surveillance, phagocytosis, and inflammation: How never-resting microglia influence adult hippocampal neurogenesis. Neural Plast. 2014, 2014, 610343. [Google Scholar] [CrossRef]
- Gehrmann, J.; Banati, R.B.; Kreutzberg, G.W. Microglia in the immune surveillance of the brain: Human microglia constitutively express HLA-DR molecules. J. Neuroimmunol. 1993, 48, 189–198. [Google Scholar] [CrossRef] [PubMed]
- Broggi, A.; Granucci, F. Microbe- and danger-induced inflammation. Mol. Immunol. 2015, 63, 127–133. [Google Scholar] [CrossRef]
- 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]
- Scheiblich, H.; Bousset, L.; Schwartz, S.; Griep, A.; Latz, E.; Melki, R.; Heneka, M.T. Microglial NLRP3 Inflammasome Activation upon TLR2 and TLR5 Ligation by Distinct α-Synuclein Assemblies. J. Immunol. 2021, 207, 2143–2154. [Google Scholar] [CrossRef]
- Dzamko, N.; Gysbers, A.; Perera, G.; Bahar, A.; Shankar, A.; Gao, J.; Fu, Y.; Halliday, G.M. Toll-like receptor 2 is increased in neurons in Parkinson’s disease brain and may contribute to alpha-synuclein pathology. Acta Neuropathol. 2017, 133, 303–319. [Google Scholar] [CrossRef]
- Stefanova, N.; Fellner, L.; Reindl, M.; Masliah, E.; Poewe, W.; Wenning, G.K. Toll-like receptor 4 promotes α-synuclein clearance and survival of nigral dopaminergic neurons. Am. J. Pathol. 2011, 179, 954–963. [Google Scholar] [CrossRef]
- Cao, S.; Standaert, D.G.; Harms, A.S. The gamma chain subunit of Fc receptors is required for alpha-synuclein-induced pro-inflammatory signaling in microglia. J. Neuroinflammation 2012, 9, 259. [Google Scholar] [CrossRef] [PubMed]
- Smith, K.G.; Clatworthy, M.R. FcgammaRIIB in autoimmunity and infection: Evolutionary and therapeutic implications. Nat. Rev. Immunol. 2010, 10, 328–343. [Google Scholar] [CrossRef] [PubMed]
- Su, X.; Maguire-Zeiss, K.A.; Giuliano, R.; Prifti, L.; Venkatesh, K.; Federoff, H.J. Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol. Aging 2008, 29, 1690–1701. [Google Scholar] [CrossRef]
- Bennett, N.K.; Chmielowski, R.; Abdelhamid, D.S.; Faig, J.J.; Francis, N.; Baum, J.; Pang, Z.P.; Uhrich, K.E.; Moghe, P.V. Polymer brain-nanotherapeutics for multipronged inhibition of microglial α-synuclein aggregation, activation, and neurotoxicity. Biomaterials 2016, 111, 179–189. [Google Scholar] [CrossRef]
- 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]
- Zhang, W.; Dallas, S.; Zhang, D.; Guo, J.P.; Pang, H.; Wilson, B.; Miller, D.S.; Chen, B.; Zhang, W.; McGeer, P.L.; et al. Microglial PHOX and Mac-1 are essential to the enhanced dopaminergic neurodegeneration elicited by A30P and A53T mutant alpha-synuclein. Glia 2007, 55, 1178–1188. [Google Scholar] [CrossRef] [PubMed]
- Gasparotto, J.; Somensi, N.; Girardi, C.S.; Bittencourt, R.R.; de Oliveira, L.M.; Hoefel, L.P.; Scheibel, I.M.; Peixoto, D.O.; Moreira, J.C.F.; Outeiro, T.F.; et al. Is it all the RAGE? Defining the role of the receptor for advanced glycation end products in Parkinson’s disease. J. Neurochem. 2024, 168, 1608–1624. [Google Scholar] [CrossRef] [PubMed]
- Bido, S.; Muggeo, S.; Massimino, L.; Marzi, M.J.; Giannelli, S.G.; Melacini, E.; Nannoni, M.; Gambarè, D.; Bellini, E.; Ordazzo, G.; et al. Author Correction: Microglia-specific overexpression of α-synuclein leads to severe dopaminergic neurodegeneration by phagocytic exhaustion and oxidative toxicity. Nat. Commun. 2021, 12, 7359. [Google Scholar] [CrossRef]
- Long, H.; Zhang, S.; Zeng, S.; Tong, Y.; Liu, J.; Liu, C.; Li, D. Interaction of RAGE with α-synuclein fibrils mediates inflammatory response of microglia. Cell Rep. 2022, 40, 111401. [Google Scholar] [CrossRef]
- Yin, S.; Chi, X.; Wan, F.; Li, Y.; Zhou, Q.; Kou, L.; Sun, Y.; Wu, J.; Zou, W.; Wang, Y.; et al. TREM2 signaling in Parkinson’s disease: Regulation of microglial function and α-synuclein pathology. Int. Immunopharmacol. 2024, 143, 113446. [Google Scholar] [CrossRef]
- Guo, Y.; Wei, X.; Yan, H.; Qin, Y.; Yan, S.; Liu, J.; Zhao, Y.; Jiang, F.; Lou, H. TREM2 deficiency aggravates α-synuclein-induced neurodegeneration and neuroinflammation in Parkinson’s disease models. FASEB J. 2019, 33, 12164–12174. [Google Scholar] [CrossRef]
- Huang, P.; Zhang, Z.; Zhang, P.; Feng, J.; Xie, J.; Zheng, Y.; Liang, X.; Zhu, B.; Chen, Z.; Feng, S.; et al. TREM2 Deficiency Aggravates NLRP3 Inflammasome Activation and Pyroptosis in MPTP-Induced Parkinson’s Disease Mice and LPS-Induced BV2 Cells. Mol. Neurobiol. 2024, 61, 2590–2605. [Google Scholar] [CrossRef]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
- Turola, E.; Furlan, R.; Bianco, F.; Matteoli, M.; Verderio, C. Microglial microvesicle secretion and intercellular signaling. Front. Physiol. 2012, 3, 149. [Google Scholar] [CrossRef]
- Shi, M.; Liu, C.; Cook, T.J.; Bullock, K.M.; Zhao, Y.; Ginghina, C.; Li, Y.; Aro, P.; Dator, R.; He, C.; et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014, 128, 639–650. [Google Scholar] [CrossRef]
- Chang, C.; Lang, H.; Geng, N.; Wang, J.; Li, N.; Wang, X. Exosomes of BV-2 cells induced by alpha-synuclein: Important mediator of neurodegeneration in PD. Neurosci. Lett. 2013, 548, 190–195. [Google Scholar] [CrossRef]
- Alvarez-Erviti, L.; Seow, Y.; Schapira, A.H.; Gardiner, C.; Sargent, I.L.; Wood, M.J.; Cooper, J.M. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol. Dis. 2011, 42, 360–367. [Google Scholar] [CrossRef]
- Vacchi, E.; Burrello, J.; Di Silvestre, D.; Burrello, A.; Bolis, S.; Mauri, P.; Vassalli, G.; Cereda, C.W.; Farina, C.; Barile, L.; et al. Immune profiling of plasma-derived extracellular vesicles identifies Parkinson disease. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e866. [Google Scholar] [CrossRef]
- Tarasiuk, O.; Scuteri, A. Role of Tunneling Nanotubes in the Nervous System. Int. J. Mol. Sci. 2022, 23, 12545. [Google Scholar] [CrossRef]
- Dieriks, B.V.; Park, T.I.; Fourie, C.; Faull, R.L.; Dragunow, M.; Curtis, M.A. α-synuclein transfer through tunneling nanotubes occurs in SH-SY5Y cells and primary brain pericytes from Parkinson’s disease patients. Sci. Rep. 2017, 7, 42984. [Google Scholar] [CrossRef] [PubMed]
- Dilsizoglu Senol, A.; Samarani, M.; Syan, S.; Guardia, C.M.; Nonaka, T.; Liv, N.; Latour-Lambert, P.; Hasegawa, M.; Klumperman, J.; Bonifacino, J.S.; et al. α-Synuclein fibrils subvert lysosome structure and function for the propagation of protein misfolding between cells through tunneling nanotubes. PLoS Biol. 2021, 19, e3001287. [Google Scholar] [CrossRef] [PubMed]
- Scheiblich, H.; Dansokho, C.; Mercan, D.; Schmidt, S.V.; Bousset, L.; Wischhof, L.; Eikens, F.; Odainic, A.; Spitzer, J.; Griep, A.; et al. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell 2021, 184, 5089–5106.e5021. [Google Scholar] [CrossRef] [PubMed]
- Arcuri, C.; Mecca, C.; Bianchi, R.; Giambanco, I.; Donato, R. The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS. Front. Mol. Neurosci. 2017, 10, 191. [Google Scholar] [CrossRef] [PubMed]
- Daniele, S.G.; Béraud, D.; Davenport, C.; Cheng, K.; Yin, H.; Maguire-Zeiss, K.A. Activation of MyD88-dependent TLR1/2 signaling by misfolded α-synuclein, a protein linked to neurodegenerative disorders. Sci. Signal 2015, 8, ra45. [Google Scholar] [CrossRef]
- Anderson, F.L.; Biggs, K.E.; Rankin, B.E.; Havrda, M.C. NLRP3 inflammasome in neurodegenerative disease. Transl. Res. 2023, 252, 21–33. [Google Scholar] [CrossRef]
- Holbrook, J.A.; Jarosz-Griffiths, H.H.; Caseley, E.; Lara-Reyna, S.; Poulter, J.A.; Williams-Gray, C.H.; Peckham, D.; McDermott, M.F. Neurodegenerative Disease and the NLRP3 Inflammasome. Front. Pharmacol. 2021, 12, 643254. [Google Scholar] [CrossRef]
- Broz, P.; Pelegrín, P.; Shao, F. The gasdermins, a protein family executing cell death and inflammation. Nat. Rev. Immunol. 2020, 20, 143–157. [Google Scholar] [CrossRef]
- Weir, A.; Vince, J.E. No longer married to inflammasome signaling: The diverse interacting pathways leading to pyroptotic cell death. Biochem. J. 2022, 479, 1083–1102. [Google Scholar] [CrossRef]
- De Simone, M.; Choucha, A.; Ciaglia, E.; Conti, V.; Pecoraro, G.; Santurro, A.; Puca, A.A.; Cascella, M.; Iaconetta, G. Discogenic Low Back Pain: Anatomic and Pathophysiologic Characterization, Clinical Evaluation, Biomarkers, AI, and Treatment Options. J. Clin. Med. 2024, 13, 5915. [Google Scholar] [CrossRef]
- Hrabos, D.; Poggiolini, I.; Civitelli, L.; Galli, E.; Esapa, C.; Saarma, M.; Lindholm, P.; Parkkinen, L. Unfolded protein response markers Grp78 and eIF2alpha are upregulated with increasing alpha-synuclein levels in Lewy body disease. Neuropathol. Appl. Neurobiol. 2024, 50, e12999. [Google Scholar] [CrossRef]
- Chavarría, C.; Ivagnes, R.; Souza, J.M. Extracellular Alpha-Synuclein: Mechanisms for Glial Cell Internalization and Activation. Biomolecules 2022, 12, 655. [Google Scholar] [CrossRef] [PubMed]
- Peña-Martinez, C.; Rickman, A.D.; Heckmann, B.L. Beyond autophagy: LC3-associated phagocytosis and endocytosis. Sci. Adv. 2022, 8, eabn1702. [Google Scholar] [CrossRef]
- Janda, E.; Boi, L.; Carta, A.R. Microglial Phagocytosis and Its Regulation: A Therapeutic Target in Parkinson’s Disease? Front. Mol. Neurosci. 2018, 11, 144. [Google Scholar] [CrossRef]
- Webb, J.L.; Ravikumar, B.; Atkins, J.; Skepper, J.N.; Rubinsztein, D.C. Alpha-Synuclein is degraded by both autophagy and the proteasome. J. Biol. Chem. 2003, 278, 25009–25013. [Google Scholar] [CrossRef]
- Tanida, I. Autophagosome formation and molecular mechanism of autophagy. Antioxid. Redox Signal. 2011, 14, 2201–2214. [Google Scholar] [CrossRef]
- Wong, S.W.; Sil, P.; Martinez, J. Rubicon: LC3-associated phagocytosis and beyond. FEBS J. 2018, 285, 1379–1388. [Google Scholar] [CrossRef]
- Heckmann, B.L.; Teubner, B.J.W.; Tummers, B.; Boada-Romero, E.; Harris, L.; Yang, M.; Guy, C.S.; Zakharenko, S.S.; Green, D.R. LC3-Associated Endocytosis Facilitates β-Amyloid Clearance and Mitigates Neurodegeneration in Murine Alzheimer’s Disease. Cell 2020, 183, 1733–1734. [Google Scholar] [CrossRef]
- Mizushima, N.; Yoshimori, T.; Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell Dev. Biol. 2011, 27, 107–132. [Google Scholar] [CrossRef]
- Heckmann, B.L.; Green, D.R. LC3-associated phagocytosis at a glance. J. Cell Sci. 2019, 132, jcs222984. [Google Scholar] [CrossRef]
- Heckmann, B.L.; Boada-Romero, E.; Cunha, L.D.; Magne, J.; Green, D.R. LC3-Associated Phagocytosis and Inflammation. J. Mol. Biol. 2017, 429, 3561–3576. [Google Scholar] [CrossRef]
- Dodson, M.; Darley-Usmar, V.; Zhang, J. Cellular metabolic and autophagic pathways: Traffic control by redox signaling. Free Radic. Biol. Med. 2013, 63, 207–221. [Google Scholar] [CrossRef] [PubMed]
- Rojanathammanee, L.; Murphy, E.J.; Combs, C.K. Expression of mutant alpha-synuclein modulates microglial phenotype in vitro. J. Neuroinflammation 2011, 8, 44. [Google Scholar] [CrossRef]
- Roodveldt, C.; Labrador-Garrido, A.; Gonzalez-Rey, E.; Fernandez-Montesinos, R.; Caro, M.; Lachaud, C.C.; Waudby, C.A.; Delgado, M.; Dobson, C.M.; Pozo, D. Glial innate immunity generated by non-aggregated alpha-synuclein in mouse: Differences between wild-type and Parkinson’s disease-linked mutants. PLoS ONE 2010, 5, e13481. [Google Scholar] [CrossRef]
- Park, J.Y.; Paik, S.R.; Jou, I.; Park, S.M. Microglial phagocytosis is enhanced by monomeric alpha-synuclein, not aggregated alpha-synuclein: Implications for Parkinson’s disease. Glia 2008, 56, 1215–1223. [Google Scholar] [CrossRef]
- Bliederhaeuser, C.; Grozdanov, V.; Speidel, A.; Zondler, L.; Ruf, W.P.; Bayer, H.; Kiechle, M.; Feiler, M.S.; Freischmidt, A.; Brenner, D.; et al. Age-dependent defects of alpha-synuclein oligomer uptake in microglia and monocytes. Acta Neuropathol. 2016, 131, 379–391. [Google Scholar] [CrossRef]
- Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef]
- Yue, M.; Hinkle, K.M.; Davies, P.; Trushina, E.; Fiesel, F.C.; Christenson, T.A.; Schroeder, A.S.; Zhang, L.; Bowles, E.; Behrouz, B.; et al. Progressive dopaminergic alterations and mitochondrial abnormalities in LRRK2 G2019S knock-in mice. Neurobiol. Dis. 2015, 78, 172–195. [Google Scholar] [CrossRef]
- Plowey, E.D.; Cherra, S.J., III; Liu, Y.J.; Chu, C.T. Role of autophagy in G2019S-LRRK2-associated neurite shortening in differentiated SH-SY5Y cells. J. Neurochem. 2008, 105, 1048–1056. [Google Scholar] [CrossRef]
- Magalhaes, J.; Gegg, M.E.; Migdalska-Richards, A.; Doherty, M.K.; Whitfield, P.D.; Schapira, A.H. Autophagic lysosome reformation dysfunction in glucocerebrosidase deficient cells: Relevance to Parkinson disease. Hum. Mol. Genet. 2016, 25, 3432–3445. [Google Scholar] [CrossRef]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Tsutsumi, R.; Hori, Y.; Seki, T.; Kurauchi, Y.; Sato, M.; Oshima, M.; Hisatsune, A.; Katsuki, H. Involvement of exosomes in dopaminergic neurodegeneration by microglial activation in midbrain slice cultures. Biochem. Biophys. Res. Commun. 2019, 511, 427–433. [Google Scholar] [CrossRef]
- Hughes, C.D.; Choi, M.L.; Ryten, M.; Hopkins, L.; Drews, A.; Botía, J.A.; Iljina, M.; Rodrigues, M.; Gagliano, S.A.; Gandhi, S.; et al. Picomolar concentrations of oligomeric alpha-synuclein sensitizes TLR4 to play an initiating role in Parkinson’s disease pathogenesis. Acta Neuropathol. 2019, 137, 103–120. [Google Scholar] [CrossRef]
- Khan, A.; Johnson, R.; Wittmer, C.; Maile, M.; Tatsukawa, K.; Wong, J.L.; Gill, M.B.; Stocking, E.M.; Natala, S.R.; Paulino, A.D.; et al. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson’s disease. Brain 2021, 144, 3692–3709. [Google Scholar] [CrossRef]
- Li, Y.; Tong, Q.; Wang, Y.; Cheng, Y.; Geng, Y.; Tian, T.; Yuan, Y.; Fan, Y.; Lu, M.; Zhang, K. Phosphorylated α-synuclein deposited in Schwann cells interacting with TLR2 mediates cell damage and induces Parkinson’s disease autonomic dysfunction. Cell Death Discov. 2024, 10, 52. [Google Scholar] [CrossRef]
- Kim, C.; Ojo-Amaize, E.; Spencer, B.; Rockenstein, E.; Mante, M.; Desplats, P.; Wrasidlo, W.; Adame, A.; Nchekwube, E.; Oyemade, O.; et al. Hypoestoxide reduces neuroinflammation and α-synuclein accumulation in a mouse model of Parkinson’s disease. J. Neuroinflammation 2015, 12, 236. [Google Scholar] [CrossRef]
- Valera, E.; Mante, M.; Anderson, S.; Rockenstein, E.; Masliah, E. Lenalidomide reduces microglial activation and behavioral deficits in a transgenic model of Parkinson’s disease. J. Neuroinflammation 2015, 12, 93. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, Q.; Yu, R.; Zhang, Q.; Zhang, Z.; Li, H.; Ren, C.; Yang, R.; Niu, H. Minocycline inhibition of microglial rescues nigrostriatal dopaminergic neurodegeneration caused by mutant alpha-synuclein overexpression. Aging 2020, 12, 14232–14243. [Google Scholar] [CrossRef]
- Parashos, S.A.; Luo, S.; Biglan, K.M.; Bodis-Wollner, I.; He, B.; Liang, G.S.; Ross, G.W.; Tilley, B.C.; Shulman, L.M. Measuring disease progression in early Parkinson disease: The National Institutes of Health Exploratory Trials in Parkinson Disease (NET-PD) experience. JAMA Neurol. 2014, 71, 710–716. [Google Scholar] [CrossRef] [PubMed]
- A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006, 66, 664–671. [CrossRef]
- Cyske, Z.; Gaffke, L.; Rintz, E.; Wiśniewska, K.; Węgrzyn, G.; Pierzynowska, K. Molecular mechanisms of the ambroxol action in Gaucher disease and GBA1 mutation-associated Parkinson disease. Neurochem. Int. 2024, 178, 105774. [Google Scholar] [CrossRef]
- McNeill, A.; Magalhaes, J.; Shen, C.; Chau, K.Y.; Hughes, D.; Mehta, A.; Foltynie, T.; Cooper, J.M.; Abramov, A.Y.; Gegg, M.; et al. Ambroxol improves lysosomal biochemistry in glucocerebrosidase mutation-linked Parkinson disease cells. Brain 2014, 137, 1481–1495. [Google Scholar] [CrossRef] [PubMed]
- Mullin, S.; Smith, L.; Lee, K.; D’Souza, G.; Woodgate, P.; Elflein, J.; Hällqvist, J.; Toffoli, M.; Streeter, A.; Hosking, J.; et al. Ambroxol for the Treatment of Patients With Parkinson Disease With and Without Glucocerebrosidase Gene Mutations: A Nonrandomized, Noncontrolled Trial. JAMA Neurol. 2020, 77, 427–434. [Google Scholar] [CrossRef]
- Chen, Y.; Xu, S.; Wang, N.; Ma, Q.; Peng, P.; Yu, Y.; Zhang, L.; Ying, Z.; Wang, H. Dynasore Suppresses mTORC1 Activity and Induces Autophagy to Regulate the Clearance of Protein Aggregates in Neurodegenerative Diseases. Neurotox. Res. 2019, 36, 108–116. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Sung, K.W.; Bae, E.J.; Yoon, D.; Kim, D.; Lee, J.S.; Park, D.H.; Park, D.Y.; Mun, S.R.; Kwon, S.C.; et al. Targeted degradation of ⍺-synuclein aggregates in Parkinson’s disease using the AUTOTAC technology. Mol. Neurodegener. 2023, 18, 41. [Google Scholar] [CrossRef] [PubMed]
- Schapira, A.H.; Tolosa, E. Molecular and clinical prodrome of Parkinson disease: Implications for treatment. Nat. Rev. Neurol. 2010, 6, 309–317. [Google Scholar] [CrossRef] [PubMed]
- Lin, M.K.; Farrer, M.J. Genetics and genomics of Parkinson’s disease. Genome Med. 2014, 6, 48. [Google Scholar] [CrossRef]
- Blauwendraat, C.; Nalls, M.A.; Singleton, A.B. The genetic architecture of Parkinson’s disease. Lancet Neurol. 2020, 19, 170–178. [Google Scholar] [CrossRef]
- Straccia, G.; Colucci, F.; Eleopra, R.; Cilia, R. Precision Medicine in Parkinson’s Disease: From Genetic Risk Signals to Personalized Therapy. Brain Sci. 2022, 12, 1308. [Google Scholar] [CrossRef]
- Reed, X.; Schumacher-Schuh, A.; Hu, J.; Bandres-Ciga, S. Advancing Personalized Medicine in Common Forms of Parkinson’s Disease through Genetics: Current Therapeutics and the Future of Individualized Management. J. Pers. Med. 2021, 11, 169. [Google Scholar] [CrossRef]
- Yoon, H.H.; Ye, S.; Lim, S.; Jo, A.; Lee, H.; Hong, F.; Lee, S.E.; Oh, S.J.; Kim, N.R.; Kim, K.; et al. CRISPR-Cas9 Gene Editing Protects from the A53T-SNCA Overexpression-Induced Pathology of Parkinson’s Disease In Vivo. CRISPR J. 2022, 5, 95–108. [Google Scholar] [CrossRef]
- Kantor, B.; Tagliafierro, L.; Gu, J.; Zamora, M.E.; Ilich, E.; Grenier, C.; Huang, Z.Y.; Murphy, S.; Chiba-Falek, O. Downregulation of SNCA Expression by Targeted Editing of DNA Methylation: A Potential Strategy for Precision Therapy in PD. Mol. Ther. 2018, 26, 2638–2649. [Google Scholar] [CrossRef] [PubMed]
- Thome, A.D.; Harms, A.S.; Volpicelli-Daley, L.A.; Standaert, D.G. microRNA-155 Regulates Alpha-Synuclein-Induced Inflammatory Responses in Models of Parkinson Disease. J. Neurosci. 2016, 36, 2383–2390. [Google Scholar] [CrossRef]
- Zhang, J.; Dongwei, Z.; Zhang, Z.; Xinhui, Q.; Kunwang, B.; Guohui, L.; Jian, D. miR-let-7a suppresses α-Synuclein-induced microglia inflammation through targeting STAT3 in Parkinson’s disease. Biochem. Biophys. Res. Commun. 2019, 519, 740–746. [Google Scholar] [CrossRef] [PubMed]
- Hudry, E.; Vandenberghe, L.H. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 2019, 101, 839–862. [Google Scholar] [CrossRef]
- Singh, A.; Sen, D. Therapeutic Value of Adeno Associated Virus as a Gene Therapy Vector for Parkinson’s Disease—A Focused Review. Curr. Gene Ther. 2016, 16, 278–286. [Google Scholar] [CrossRef]
- Hitti, F.L.; Yang, A.I.; Gonzalez-Alegre, P.; Baltuch, G.H. Human gene therapy approaches for the treatment of Parkinson’s disease: An overview of current and completed clinical trials. Park. Relat. Disord. 2019, 66, 16–24. [Google Scholar] [CrossRef]
- Nam, Y.; Moon, G.J.; Kim, S.R. Therapeutic Potential of AAV1-Rheb(S16H) Transduction against Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 3064. [Google Scholar] [CrossRef]
- Ball, J.B.; Frank, M.G.; Green-Fulgham, S.M.; Watkins, L.R. Use of adeno-associated viruses for transgenic modulation of microglia structure and function: A review of technical considerations and challenges. Brain. Behav. Immun. 2024, 118, 368–379. [Google Scholar] [CrossRef]
- Lin, R.; Zhou, Y.; Yan, T.; Wang, R.; Li, H.; Wu, Z.; Zhang, X.; Zhou, X.; Zhao, F.; Zhang, L.; et al. Directed evolution of adeno-associated virus for efficient gene delivery to microglia. Nat. Methods 2022, 19, 976–985. [Google Scholar] [CrossRef]
- Shah, S.; Wong, L.M.; Ellis, K.; Bodnar, B.; Saribas, S.; Ting, J.; Wei, Z.; Tang, Y.; Wang, X.; Wang, H.; et al. Microglia-Specific Promoter Activities of HEXB Gene. Front. Cell. Neurosci. 2022, 16, 808598. [Google Scholar] [CrossRef] [PubMed]
- Okada, Y.; Hosoi, N.; Matsuzaki, Y.; Fukai, Y.; Hiraga, A.; Nakai, J.; Nitta, K.; Shinohara, Y.; Konno, A.; Hirai, H. Development of microglia-targeting adeno-associated viral vectors as tools to study microglial behavior in vivo. Commun. Biol. 2022, 5, 1224. [Google Scholar] [CrossRef] [PubMed]
- O’Carroll, S.J.; Cook, W.H.; Young, D. AAV Targeting of Glial Cell Types in the Central and Peripheral Nervous System and Relevance to Human Gene Therapy. Front. Mol. Neurosci. 2020, 13, 618020. [Google Scholar] [CrossRef]
- Maes, M.E.; Colombo, G.; Schulz, R.; Siegert, S. Targeting microglia with lentivirus and AAV: Recent advances and remaining challenges. Neurosci. Lett. 2019, 707, 134310. [Google Scholar] [CrossRef] [PubMed]
- Rosario, A.M.; Cruz, P.E.; Ceballos-Diaz, C.; Strickland, M.R.; Siemienski, Z.; Pardo, M.; Schob, K.L.; Li, A.; Aslanidi, G.V.; Srivastava, A.; et al. Microglia-specific targeting by novel capsid-modified AAV6 vectors. Mol. Ther. Methods Clin. Dev. 2016, 3, 16026. [Google Scholar] [CrossRef]
- Xue, X.; Duan, R.; Zheng, G.; Chen, H.; Zhang, W.; Shi, L. Translocator protein (18 kDa) regulates the microglial phenotype in Parkinson’s disease through P47. Bioengineered 2022, 13, 11061–11071. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Iannaccone, S.; Cerami, C.; Alessio, M.; Garibotto, V.; Panzacchi, A.; Olivieri, S.; Gelsomino, G.; Moresco, R.; Perani, D. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Park. Relat. Disord. 2013, 19, 47–52. [Google Scholar] [CrossRef]
- Fezeu, F.; Jbara, O.F.; Jbarah, A.; Choucha, A.; De Maria, L.; Ciaglia, E.; De Simone, M.; Samnick, S. PET imaging for a very early detection of rapid eye movement sleep behaviour disorder and Parkinson’s disease—A model-based cost-effectiveness analysis. Clin. Neurol. Neurosurg. 2024, 243, 108404. [Google Scholar] [CrossRef]
- Frison, M.; Faccenda, D.; Abeti, R.; Rigon, M.; Strobbe, D.; England-Rendon, B.S.; Cash, D.; Barnes, K.; Sadeghian, M.; Sajic, M.; et al. The translocator protein (TSPO) is prodromal to mitophagy loss in neurotoxicity. Mol. Psychiatry 2021, 26, 2721–2739. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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
Eo, H.; Kim, S.; Jung, U.J.; Kim, S.R. Alpha-Synuclein and Microglia in Parkinson’s Disease: From Pathogenesis to Therapeutic Prospects. J. Clin. Med. 2024, 13, 7243. https://doi.org/10.3390/jcm13237243
Eo H, Kim S, Jung UJ, Kim SR. Alpha-Synuclein and Microglia in Parkinson’s Disease: From Pathogenesis to Therapeutic Prospects. Journal of Clinical Medicine. 2024; 13(23):7243. https://doi.org/10.3390/jcm13237243
Chicago/Turabian StyleEo, Hyemi, Sehwan Kim, Un Ju Jung, and Sang Ryong Kim. 2024. "Alpha-Synuclein and Microglia in Parkinson’s Disease: From Pathogenesis to Therapeutic Prospects" Journal of Clinical Medicine 13, no. 23: 7243. https://doi.org/10.3390/jcm13237243
APA StyleEo, H., Kim, S., Jung, U. J., & Kim, S. R. (2024). Alpha-Synuclein and Microglia in Parkinson’s Disease: From Pathogenesis to Therapeutic Prospects. Journal of Clinical Medicine, 13(23), 7243. https://doi.org/10.3390/jcm13237243