Neurodegenerative disorders present a significant global health challenge. Neuroinflammation, which is a frequent occurrence of these diseases, may serve as an initial trigger. Emerging evidence suggests that microglia play a pivotal role as mediators of inflammation in the central nervous system. Disordered activation skews predominantly towards the M1 phenotype, which leads to neurotoxicity and neuroinflammation. Identifying the triggers of microglial activation is crucial to refine the therapeutic strategies for neurodegenerative diseases. Based on epidemiological, biological, and functional data, we observed a strong correlation between chronic microbial infection and microglial activation. This review outlines three potential inter-organ communication routes for infectious microbes in microglial activation—the gut-brain axis, the lung-brain axis, and the nose-brain axis—thus emphasizing the interaction between microglia and inflammation and proving that neuroinflammation is a significant factor in pathogen-induced neurodegenerative diseases. Collectively, we propose that therapies which target relevant inflammatory mediators, along with antibiotics that target neuropathogens, could potentially alleviate neuroinflammation and treat neurodegenerative diseases.
Citation: Tianshi Jiang, Kaili Zhu, Guangli Kang, Guojun Wu, Yurong Tan. Neuroinflammation in neurodegenerative disorders: Activation of microglia by microbial infection[J]. AIMS Molecular Science, 2025, 12(3): 198-215. doi: 10.3934/molsci.2025013
Neurodegenerative disorders present a significant global health challenge. Neuroinflammation, which is a frequent occurrence of these diseases, may serve as an initial trigger. Emerging evidence suggests that microglia play a pivotal role as mediators of inflammation in the central nervous system. Disordered activation skews predominantly towards the M1 phenotype, which leads to neurotoxicity and neuroinflammation. Identifying the triggers of microglial activation is crucial to refine the therapeutic strategies for neurodegenerative diseases. Based on epidemiological, biological, and functional data, we observed a strong correlation between chronic microbial infection and microglial activation. This review outlines three potential inter-organ communication routes for infectious microbes in microglial activation—the gut-brain axis, the lung-brain axis, and the nose-brain axis—thus emphasizing the interaction between microglia and inflammation and proving that neuroinflammation is a significant factor in pathogen-induced neurodegenerative diseases. Collectively, we propose that therapies which target relevant inflammatory mediators, along with antibiotics that target neuropathogens, could potentially alleviate neuroinflammation and treat neurodegenerative diseases.
| [1] |
Borst K, Dumas AA, Prinz M (2021) Microglia: Immune and non-immune functions. Immunity 54: 2194-2208. https://doi.org/10.1016/j.immuni.2021.09.014
|
| [2] |
Zhang W, Xiao D, Mao Q, et al. (2023) Role of neuroinflammation in neurodegeneration development. Sig Transduct Target Ther 8: 267. https://doi.org/10.1038/s41392-023-01486-5
|
| [3] |
Heneka MT, Carson MJ, El Khoury J, et al. (2015) Neuroinflammation in Alzheimer's disease. Lancet Neurol 14: 388-405. https://doi.org/10.1016/S1474-4422(15)70016-5
|
| [4] |
Zhou J, Wade SD, Graykowski D, et al. (2023) The neuronal pentraxin Nptx2 regulates complement activity and restrains microglia-mediated synapse loss in neurodegeneration. Sci Transl Med 15: eadf0141. https://doi.org/10.1126/scitranslmed.adf0141
|
| [5] |
Zipp F, Bittner S, Schafer DP (2023) Cytokines as emerging regulators of central nervous system synapses. Immunity 56: 914-925. https://doi.org/10.1016/j.immuni.2023.04.011
|
| [6] |
Jafari M, Schumacher AM, Snaidero N, et al. (2021) Phagocyte-mediated synapse removal in cortical neuroinflammation is promoted by local calcium accumulation. Nat Neurosci 24: 355-367. https://doi.org/10.1038/s41593-020-00780-7
|
| [7] |
Yuan J, Amin P, Ofengeim D (2019) Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci 20: 19-33. https://doi.org/10.1038/s41583-018-0093-1
|
| [8] |
Ising C, Venegas C, Zhang S, et al. (2019) NLRP3 inflammasome activation drives tau pathology. Nature 575: 669-673. https://doi.org/10.1038/s41586-019-1769-z
|
| [9] |
Zheng C, Zhou XW, Wang JZ (2016) The dual roles of cytokines in Alzheimer's disease: Update on interleukins, TNF-α, TGF-β and IFN-γ. Transl Neurodegener 5: 7. https://doi.org/10.1186/s40035-016-0054-4
|
| [10] |
McAlpine CS, Park J, Griciuc A, et al. (2021) Astrocytic interleukin-3 programs microglia and limits Alzheimer's disease. Nature 595: 701-706. https://doi.org/10.1038/s41586-021-03734-6
|
| [11] |
Gao C, Jiang J, Tan Y, et al. (2023) Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Sig Transduct Target Ther 8: 359. https://doi.org/10.1038/s41392-023-01588-0
|
| [12] |
Anderson FL, Biggs KE, Rankin BE, et al. (2023) NLRP3 inflammasome in neurodegenerative disease. Transl Res 252: 21-33. https://doi.org/10.1016/j.trsl.2022.08.006
|
| [13] |
Wendimu MY, Hooks SB (2022) Microglia phenotypes in aging and neurodegenerative diseases. Cells 11: 2091. https://doi.org/10.3390/cells11132091
|
| [14] |
Zhang Z, Zheng X, Zhang X, et al. (2020) Aging alters Hv1-mediated microglial polarization and enhances neuroinflammation after peripheral surgery. CNS Neurosci Ther 26: 374-384. https://doi.org/10.1111/cns.13271
|
| [15] |
Marschallinger J, Iram T, Zardeneta M, et al. (2020) Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat Neurosci 23: 194-208. https://doi.org/10.1038/s41593-019-0566-1
|
| [16] |
Aguzzi A, Barres BA, Bennett ML (2013) Microglia: Scapegoat, saboteur, or something else?. Science 339: 156-161. https://doi.org/10.1126/science.1227901
|
| [17] |
Gulen MF, Samson N, Keller A (2023) cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature 620: 374-380. https://doi.org/10.1038/s41586-023-06373-1
|
| [18] |
Guo M, Wang J, Zhao Y, et al. (2020) Microglial exosomes facilitate α-synuclein transmission in Parkinson's disease. Brain 143: 1476-1497. https://doi.org/10.1093/brain/awaa090
|
| [19] |
Wolf SA, Boddeke HWGM, Kettenmann H (2017) Microglia in physiology and disease. Annu Rev Physiol 79: 619-643. https://doi.org/10.1146/annurev-physiol-022516-034406
|
| [20] |
Terreros-Roncal J, Moreno-Jiménez EP, Flor-García M, et al. (2021) Impact of neurodegenerative diseases on human adult hippocampal neurogenesis. Science 374: 1106-1113. https://doi.org/10.1126/science.abl5163
|
| [21] |
Dong-Chen X, Yong C, Yang X, et al. (2023) Signaling pathways in Parkinson's disease: Molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 8: 73. https://doi.org/10.1038/s41392-023-01353-3
|
| [22] |
Wu X, Zhang H, Qi W, et al. (2018) Nicotine promotes atherosclerosis via ROS-NLRP3-mediated endothelial cell pyroptosis. Cell Death Dis 9: 171. https://doi.org/10.1038/s41419-017-0257-3
|
| [23] | Lei L, Wang R, Pan Y, et al. (2021) Mangiferin inhibited neuroinflammation through regulating microglial polarization and suppressing NF-κB, NLRP3 pathway. Chin J Nat Med 19: 112-119. https://doi.org/10.1016/S1875-5364(21)60012-2 |
| [24] |
Hoffmann FS, Hofereiter J, Rübsamen H, et al. (2015) Fingolimod induces neuroprotective factors in human astrocytes. J Neuroinflammation 12: 184. https://doi.org/10.1186/s12974-015-0393-6
|
| [25] |
Chithanathan K, Jürgenson M, Guha M, et al. (2022) Paradoxical attenuation of neuroinflammatory response upon LPS challenge in miR-146b deficient mice. Front Immunol 13: 996415. https://doi.org/10.3389/fimmu.2022.996415
|
| [26] |
Deczkowska A, Amit I, Schwartz M (2018) Microglial immune checkpoint mechanisms. Nat Neurosci 21: 779-786. https://doi.org/10.1038/s41593-018-0145-x
|
| [27] |
Brown GC (2024) Cell death by phagocytosis. Nat Rev Immunol 24: 91-102. https://doi.org/10.17863/CAM.100711
|
| [28] |
Vuong HE, Yano JM, Fung TC, et al. (2017) The microbiome and host behavior. Annu Rev Neurosci 40: 21-49. https://doi.org/10.1146/annurev-neuro-072116-031347
|
| [29] |
Caballero-Flores G, Pickard JM, Núñez G (2023) Microbiota-mediated colonization resistance: Mechanisms and regulation. Nat Rev Microbiol 21: 347-360. https://doi.org/10.1038/s41579-022-00833-7
|
| [30] |
Loh JS, Mak WQ, Tan LKS, et al. (2024) Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 9: 37. https://doi.org/10.1038/s41392-024-01743-1
|
| [31] |
Miyauchi E, Shimokawa C, Steimle A, et al. (2023) The impact of the gut microbiome on extra-intestinal autoimmune diseases. Nat Rev Immunol 23: 9-23. https://doi.org/10.1038/s41577-022-00727-y
|
| [32] |
Almeida A, Mitchell AL, Boland M, et al. (2019) A new genomic blueprint of the human gut microbiota. Nature 568: 99-504. https://doi.org/10.1038/s41586-019-0965-1
|
| [33] |
Lynch SV, Pedersen O (2016) The human intestinal microbiome in health and disease. N Engl J Med 375: 2369-2379. https://doi.org/10.1056/NEJMra1600266
|
| [34] |
Erny D, de Angelis ALH, Jaitin D, et al. (2015) Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18: 965-977. https://doi.org/10.1038/nn.4030
|
| [35] |
Cook J, Prinz M (2022) Regulation of microglial physiology by the microbiota. Gut Microbes 14: 2125739. https://doi.org/10.1080/19490976.2022.2125739
|
| [36] |
Zhou R, Qian S, Cho WCS, et al. (2022) Microbiota-microglia connections in age-related cognition decline. Aging Cell 21: e13599. https://doi.org/10.1111/acel.13599
|
| [37] |
Abdel-Haq R, Schlachetzki JCM, Glass CK, et al. (2019) Microbiome-microglia connections via the gut-brain axis. J Exp Med 216: 41-59. https://doi.org/10.1084/jem.20180794
|
| [38] |
Mossad O, Erny D (2020) The microbiota-microglia axis in central nervous system disorders. Brain Pathol 30: 1159-1177. https://doi.org/10.1111/bpa.12908
|
| [39] |
Erny D, Dokalis N, Mezö C, et al. (2021) Microbiota-derived acetate enables the metabolic fitness of the brain innate immune system during health and disease. Cell Metab 33: 2260-2276.e7. https://doi.org/10.1016/j.cmet.2021.10.010
|
| [40] |
Liu X, Li X, Xia B, et al. (2021) High-fiber diet mitigates maternal obesity-induced cognitive and social dysfunction in the offspring via gut-brain axis. Cell Metab 33: 923-938.e6. https://doi.org/10.1016/j.cmet.2021.02.002
|
| [41] |
Meng Y, Qiu X, Tang Z, et al. (2023) Lactobacillus paracasei L9 affects disease progression in experimental autoimmune neuritis by regulating intestinal flora structure and arginine metabolism. Neuroinflamm 20: 122. https://doi.org/10.1186/s12974-023-02808-8
|
| [42] |
Cryan JF, O'Riordan KJ, Cowan CSM, et al. (2019) The microbiota-gut-brain axis. Physiol Rev 99: 1877-2013. https://doi.org/10.1152/physrev.00018.2018
|
| [43] |
Man WH, de Steenhuijsen Piters WAA, Bogaert D (2017) The microbiota of the respiratory tract: Gatekeeper to respiratory health. Nat Rev Microbiol 15: 259-270. https://doi.org/10.1038/nrmicro.2017.14
|
| [44] |
Sankowski R, Ahmari J, Mezö C, et al. (2021) Commensal microbiota divergently affect myeloid subsets in the mammalian central nervous system during homeostasis and disease. EMBO J 40: e108605. https://doi.org/10.15252/embj.2021108605
|
| [45] |
Pokusaeva K, Johnson C, Luk B, et al. (2017) GABA-producing Bifidobacterium dentium modulates visceral sensitivity in the intestine. Neurogastroenterol Motil 29: e12904. https://doi.org/10.1111/nmo.12904
|
| [46] |
Yano JM, Yu K, Donaldson GP, et al. (2015) Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161: 264-276. https://doi.org/10.1016/j.cell.2015.02.047
|
| [47] |
Teng Y, Mu J, Xu F, et al. (2022) Gut bacterial isoamylamine promotes age-related cognitive dysfunction by promoting microglial cell death. Cell Host Microbe 30: 944-960.e8. https://doi.org/10.1016/j.chom.2022.05.005
|
| [48] |
Lamers MM, Haagmans BL (2022) SARS-CoV-2 pathogenesis. Nat Rev Microbiol 20: 270-284. https://doi.org/10.1038/s41579-022-00713-0
|
| [49] |
Scher JU, Joshua V, Artacho A, et al. (2016) The lung microbiota in early rheumatoid arthritis and autoimmunity. Microbiome 4: 60. https://doi.org/10.1186/s40168-016-0206-x
|
| [50] |
Kerr N, de Rivero Vaccari JP, Dietrich WD, et al. (2020) Neural-respiratory inflammasome axis in traumatic brain injury. Exp Neurol 323: 113080. https://doi.org/10.1016/j.expneurol.2019.113080
|
| [51] |
Forés-Martos J, Catalá-López F, Sánchez-Valle J, et al. (2019) Transcriptomic metaanalyses of autistic brains reveals shared gene expression and biological pathway abnormalities with cancer. Mol Autism 10: 17. https://doi.org/10.1186/s13229-019-0262-8
|
| [52] |
Tran VTA, Lee LP, Cho H (2022) Neuroinflammation in neurodegeneration via microbial infections. Front Immunol 13: 907804. https://doi.org/10.3389/fimmu.2022.907804
|
| [53] |
Hosang L, Canals RC, van der Flier FJ, et al. (2022) The lung microbiome regulates brain autoimmunity. Nature 603: 138-144. https://doi.org/10.1038/s41586-022-04427-4
|
| [54] |
McNab F, Mayer-Barber K, Sher A, et al. (2015) Type I interferons in infectious disease. Nat Rev Immunol 15: 87-103. https://doi.org/10.1038/nri3787
|
| [55] |
Azzoni R, Marsland BJ (2022) The lung-brain axis: A new frontier in host-microbe interactions. Immunity 55: 589-591. https://doi.org/10.1016/j.immuni.2022.03.015
|
| [56] |
Bajinka O, Simbilyabo L, Tan Y, et al. (2022) Lung-brain axis. Crit Rev Microbiol 48: 257-269. https://doi.org/10.1080/1040841X.2021.1960483
|
| [57] |
Mao Y, Bajinka O, Tang Z, et al. (2022) Lung-brain axis: Metabolomics and pathological changes in lungs and brain of respiratory syncytial virus-infected mice. J Med Virol 94: 5885-5893. https://doi.org/10.1002/jmv.28061
|
| [58] |
Ji JJ, Sun QM, Nie DY, et al. (2021) Probiotics protect against RSV infection by modulating the microbiota-alveolar-macrophage axis. Acta Pharmacol Sin 42: 1630-1641. https://doi.org/10.1038/s41401-020-00573-5
|
| [59] |
Chen X, Wang X, Cui Z, et al. (2023) M1 microglia-derived exosomes promote activation of resting microglia and amplifies proangiogenic effects through Irf1/miR-155-5p/Socs1 axis in the retina. Int J Biol Sci 19: 1791-1812. https://doi.org/10.7150/ijbs.79784
|
| [60] |
Terui H, Yamasaki K, Wada-Irimada M, et al. (2022) Staphylococcus aureus skin colonization promotes SLE-like autoimmune inflammation via neutrophil activation and the IL-23/IL-17 axis. Sci Immunol 7: eabm9811. https://doi.org/10.1126/sciimmunol.abm9811
|
| [61] |
Jiang T, Zhu K, Kang G, et al. (2024) Infectious viruses and neurodegenerative diseases: The mitochondrial defect hypothesis. Rev Med Virol 34: e2565. https://doi.org/10.1002/rmv.2565
|
| [62] |
Rothhammer V, Mascanfroni ID, Bunse L, et al. (2016) Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med 22: 586-597. https://doi.org/10.1038/nm.4106
|
| [63] |
Giri PM, Banerjee A, Ghosal A, et al. (2024) Neuroinflammation in neurodegenerative disorders: Current knowledge and therapeutic implications. Int J Mol Sci 25: 3995. https://doi.org/10.3390/ijms25073995
|
| [64] |
Uddin MS, Yu WS, Lim LW (2021) Exploring ER stress response in cellular aging and neuroinflammation in Alzheimer's disease. Ageing Res Rev 70: 101417. https://doi.org/10.1016/j.arr.2021.101417
|
| [65] |
Benitez DP, Jiang S, Wood J, et al. (2021) Knock-in models related to Alzheimer's disease: synaptic transmission, plaques and the role of microglia. Mol Neurodegener 16: 47. https://doi.org/10.1186/s13024-021-00457-0
|
| [66] |
Tan EK, Chao YX, West A, et al. (2020) Parkinson disease and the immune system-associations, mechanisms and therapeutics. Nat Rev Neurol 16: 303-318. https://doi.org/10.1038/s41582-020-0344-4
|
| [67] |
Zhang W, Xiao D, Mao Q, Xia H (2023) Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 8: 267. https://doi.org/10.1038/s41392-023-01486-5
|
| [68] |
Tan EK, Chao YX, West A, et al. (2020) Parkinson disease and the immune system-associations, mechanisms and therapeutics. Nat Rev Neurol 16: 303-318. https://doi.org/10.1038/s41582-020-0344-4
|
| [69] |
Bajinka O, Tang Z, Mao Y, et al. (2023) Respiratory syncytial virus infection disrupts pulmonary microbiota to induce microglia phenotype shift. J Med Virol 95: e28976. https://doi.org/10.1002/jmv.28976
|
| [70] |
Dey R, Bishayi B (2020) TLR-2 neutralization potentiates microglial M1 to M2 switching by the combinatorial treatment of ciprofloxacin and dexamethasone during S. aureus infection. Neuroimmunol 344: 577262. https://doi.org/10.1016/j.jneuroim.2020.577262
|
| [71] |
Xie J, Shen Z, Anraku Y, et al. (2019) Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials 224: 119491. https://doi.org/10.1016/j.biomaterials.2019.119491
|
| [72] |
Hüttenrauch M, Ogorek I, Klafki H, et al. (2018) Glycoprotein NMB: A novel Alzheimer's disease associated marker expressed in a subset of activated microglia. Acta Neuropathol Commun 6: 108. https://doi.org/10.1186/s40478-018-0612-3
|