Generic placeholder image

Current Alzheimer Research

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Perspective

The Immunopathy of Alzheimer’s Disease: Innate or Adaptive?

Author(s): Donald F. Weaver*

Volume 20, Issue 2, 2023

Published on: 29 May, 2023

Page: [63 - 70] Pages: 8

DOI: 10.2174/1567205020666230517103312

Price: $65

conference banner
Abstract

Beyond the time-honoured targeting of protein misfolding and aggregation, Alzheimer’s disease needs new, innovative therapeutic directions. When exploring alternative druggable mechanisms, multifaceted in vitro and in vivo data demonstrate that immune system dysfunction is a pivotal driver of Alzheimer’s disease progression. In pursuing neuroimmunological targets, a major but often under-discussed consideration regards the issue of whether innate or adaptive immunity (or both) within the neuroimmune network should be the centre of focus when devising immunotherapeutic approaches to Alzheimer’s. This perspective article briefly reviews current data, concluding that while both innate and adaptive immunity contributes to the immunopathology of Alzheimer’s, the proinflammatory microglia and cytokines of innate immunity will provide higher yield targets with a greater likelihood of efficacy. Although it seems paradoxical to focus on a rapid, short-lived aspect of immunity when seeking approaches to a quintessentially chronic brain disease, accumulating evidence affords ample data to support the target-rich cascade of innate immunity for the development of much-needed new diagnostics and therapeutics.

[1]
West S, Bhugra P. Emerging drug targets for A β and tau in Alzheimer’s disease: A systematic review. Br J Clin Pharmacol 2015; 80(2): 221-34.
[http://dx.doi.org/10.1111/bcp.12621] [PMID: 25753046]
[2]
Bowirrat A. Immunosenescence and aging: Neuroinflammation is a prominent feature of Alzheimer’s Disease and is a likely contributor to neurodegenerative disease pathogenesis. J Pers Med 2022; 12(11): 1817.
[http://dx.doi.org/10.3390/jpm12111817] [PMID: 36579548]
[3]
Chauhan A, Nguyen JN. Bystanders or not? Microglia and lymphocytes in aging and stroke. Neural Regen Res 2023; 18(7): 1397-403.
[http://dx.doi.org/10.4103/1673-5374.360345] [PMID: 36571333]
[4]
Remarque EJ, Bollen ELEM, Weverling-Rijnsburger AWE, Laterveer JC, Blauw GJ, Westendorp RGJ. Patients with Alzheimer’s disease display a pro-inflammatory phenotype. Exp Gerontol 2001; 36(1): 171-6.
[http://dx.doi.org/10.1016/S0531-5565(00)00176-5] [PMID: 11162920]
[5]
Brosseron F, Krauthausen M, Kummer M, Heneka MT. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: A comparative overview. Mol Neurobiol 2014; 50(2): 534-44.
[http://dx.doi.org/10.1007/s12035-014-8657-1] [PMID: 24567119]
[6]
Dickson DW, Farlo J, Davies P, Crystal H, Fuld P, Yen SH. Alzheimer’s disease. A double-labeling immunohistochemical study of senile plaques. Am J Pathol 1988; 132(1): 86-101.
[PMID: 2456021]
[7]
Luber-Narod J, Rogers J. Immune system associated antigens expressed by cells of the human central nervous system. Neurosci Lett 1988; 94(1-2): 17-22.
[http://dx.doi.org/10.1016/0304-3940(88)90263-7] [PMID: 3266526]
[8]
Lampert-Etchells M, Pasinetti G, Finch C, Johnson S. Regional localization of cells containing complement C1q and C4 mRNAs in the frontal cortex during Alzheimer’s disease. Neurodegeneration 1993; 2: 111-21.
[9]
Zhang B, Gaiteri C, Bodea LG, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 2013; 153(3): 707-20.
[http://dx.doi.org/10.1016/j.cell.2013.03.030] [PMID: 23622250]
[10]
Thambisetty M, An Y, Nalls M, et al. Effect of complement CR1 on brain amyloid burden during aging and its modification by APOE genotype. Biol Psychiatry 2013; 73(5): 422-8.
[http://dx.doi.org/10.1016/j.biopsych.2012.08.015] [PMID: 23022416]
[11]
Naj AC, Jun G, Beecham GW, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat Genet 2011; 43(5): 436-41.
[http://dx.doi.org/10.1038/ng.801] [PMID: 21460841]
[12]
Jonsson T, Stefansson H, Steinberg S, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 2013; 368(2): 107-16.
[http://dx.doi.org/10.1056/NEJMoa1211103] [PMID: 23150908]
[13]
Lee DC, Rizer J, Hunt JB, Selenica MLB, Gordon MN, Morgan D. Review: Experimental manipulations of microglia in mouse models of Alzheimer’s pathology: Activation reduces amyloid but hastens tau pathology. Neuropathol Appl Neurobiol 2013; 39(1): 69-85.
[http://dx.doi.org/10.1111/nan.12002] [PMID: 23171029]
[14]
Meyer PF, Tremblay-Mercier J, Leoutsakos J, et al. INTREPAD: A randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. Neurology 2019; 92(18)
[http://dx.doi.org/10.1212/WNL.0000000000007232] [PMID: 30952794]
[15]
Jaturapatporn D, Isaac MGEKN, McCleery J, Tabet N. Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Libr 2012; (2): CD006378.
[http://dx.doi.org/10.1002/14651858.CD006378.pub2] [PMID: 22336816]
[16]
Kumar DKV, Choi SH, Washicosky KJ, et al. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med 2016; 8(340): 340ra72.
[http://dx.doi.org/10.1126/scitranslmed.aaf1059] [PMID: 27225182]
[17]
Chen X, Deng S, Wang W, et al. Human antimicrobial peptide LL-37 contributes to Alzheimer’s disease progression. Mol Psychiatry 2022; 27(11): 4790-9.
[http://dx.doi.org/10.1038/s41380-022-01790-6] [PMID: 36138130]
[18]
Bhaskar K, Maphis N, Xu G, et al. Microglial derived tumor necrosis factor-α drives Alzheimer’s disease-related neuronal cell cycle events. Neurobiol Dis 2014; 62: 273-85.
[http://dx.doi.org/10.1016/j.nbd.2013.10.007] [PMID: 24141019]
[19]
Fu AKY, Hung KW, Yuen MYF, et al. IL-33 ameliorates Alzheimer’s disease-like pathology and cognitive decline. Proc Natl Acad Sci 2016; 113(19): E2705-13.
[http://dx.doi.org/10.1073/pnas.1604032113] [PMID: 27091974]
[20]
Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013; 493(7434): 674-8.
[http://dx.doi.org/10.1038/nature11729] [PMID: 23254930]
[21]
Saresella M, La Rosa F, Piancone F, et al. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol Neurodegener 2016; 11(1): 23.
[http://dx.doi.org/10.1186/s13024-016-0088-1] [PMID: 26939933]
[22]
Golzari-Sorkheh M, Brown CE, Weaver DF, Reed MA. The NLRP3 inflammasome in the pathogenesis and treatment of Alzheimer’s Disease. J Alzheimers Dis 2021; 84(2): 579-98.
[http://dx.doi.org/10.3233/JAD-210660] [PMID: 34569958]
[23]
Tosto G, Reitz C. Genome-wide association studies in Alzheimer’s disease: a review. Curr Neurol Neurosci Rep 2013; 13(10): 381.
[http://dx.doi.org/10.1007/s11910-013-0381-0] [PMID: 23954969]
[24]
Ennerfelt HE, Lukens JR. The role of innate immunity in Alzheimer’s disease. Immunol Rev 2020; 297(1): 225-46.
[http://dx.doi.org/10.1111/imr.12896] [PMID: 32588460]
[25]
Huang X, Li Y, Fowler C, et al. Leukocyte surface biomarkers implicate deficits of innate immunity in sporadic Alzheimer’s disease. Alzheimers Dement 2022; alz.12813.
[http://dx.doi.org/10.1002/alz.12813] [PMID: 36349985]
[26]
Sweeney MD, Sagare AP, Zlokovic BV. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 2018; 14(3): 133-50.
[http://dx.doi.org/10.1038/nrneurol.2017.188] [PMID: 29377008]
[27]
Itagaki S, McGeer PL, Akiyama H. Presence of T-cytotoxic suppressor and leucocyte common antigen positive cells in Alzheimer’s disease brain tissue. Neurosci Lett 1988; 91(3): 259-64.
[http://dx.doi.org/10.1016/0304-3940(88)90690-8] [PMID: 2972943]
[28]
Togo T, Akiyama H, Iseki E, et al. Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases. J Neuroimmunol 2002; 124(1-2): 83-92.
[http://dx.doi.org/10.1016/S0165-5728(01)00496-9] [PMID: 11958825]
[29]
Farkas IG, Czigner A, Farkas E, et al. Beta-amyloid peptide-induced blood-brain barrier disruption facilitates T-cell entry into the rat brain. Acta Histochem 2003; 105(2): 115-25.
[http://dx.doi.org/10.1078/0065-1281-00696] [PMID: 12831163]
[30]
Ferretti MT, Merlini M, Späni C, et al. T-cell brain infiltration and immature antigen-presenting cells in transgenic models of Alzheimer’s disease-like cerebral amyloidosis. Brain Behav Immun 2016; 54: 211-25.
[http://dx.doi.org/10.1016/j.bbi.2016.02.009] [PMID: 26872418]
[31]
Laurent C, Dorothée G, Hunot S, et al. Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy. Brain 2017; 140(1): 184-200.
[http://dx.doi.org/10.1093/brain/aww270] [PMID: 27818384]
[32]
Merlini M, Kirabali T, Kulic L, Nitsch RM, Ferretti MT. Extravascular CD3+ T Cells in Brains of Alzheimer Disease Patients Correlate with Tau but Not with Amyloid Pathology: An Immunohistochemical Study. Neurodegener Dis 2018; 18(1): 49-56.
[http://dx.doi.org/10.1159/000486200] [PMID: 29402847]
[33]
Kunkle BW, Grenier-Boley B, Sims R, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ tau, immunity and lipid processing. Nat Genet 2019; 51(3): 414-30.
[http://dx.doi.org/10.1038/s41588-019-0358-2] [PMID: 30820047]
[34]
Marsh SE, Abud EM, Lakatos A, et al. The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci 2016; 113(9): E1316-25.
[http://dx.doi.org/10.1073/pnas.1525466113] [PMID: 26884167]
[35]
Späni C, Suter T, Derungs R, et al. Reduced β-amyloid pathology in an APP transgenic mouse model of Alzheimer’s disease lacking functional B and T cells. Acta Neuropathol Commun 2015; 3(1): 71.
[http://dx.doi.org/10.1186/s40478-015-0251-x] [PMID: 26558367]
[36]
Gericke C, Mallone A, Engelhardt B, Nitsch RM, Ferretti MT. Oligomeric forms of human amyloid-beta(1–42) inhibit antigen presentation. Front Immunol 2020; 11: 1029.
[http://dx.doi.org/10.3389/fimmu.2020.01029] [PMID: 32582162]
[37]
Britschgi M, Olin CE, Johns HT, et al. Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer’s disease. Proc Natl Acad Sci 2009; 106(29): 12145-50.
[http://dx.doi.org/10.1073/pnas.0904866106] [PMID: 19581601]
[38]
Giil LM, Kristoffersen EK, Vedeler CA, et al. Autoantibodies toward the angiotensin 2 type 1 receptor: A novel autoantibody in Alzheimer’s Disease. J Alzheimers Dis 2015; 47(2): 523-9.
[http://dx.doi.org/10.3233/JAD-150053] [PMID: 26401573]
[39]
Jianming W, Ling L. Autoantibodies in Alzheimer’s disease: Potential biomarkers, pathogenic roles, and therapeutic implications. J Biomed Res 2016; 30(5): 361-72.
[http://dx.doi.org/10.7555/JBR.30.20150131] [PMID: 27476881]
[40]
Mecocci P, Parnetti L, Romano G, et al. Serum anti-GFAP and anti-S100 autoantibodies in brain aging, Alzheimer’s disease and vascular dementia. J Neuroimmunol 1995; 57(1-2): 165-70.
[http://dx.doi.org/10.1016/0165-5728(94)00180-V] [PMID: 7706432]
[41]
Doss S, Wandinger KP, Hyman BT, et al. High prevalence of NMDA receptor IgA/IgM antibodies in different dementia types. Ann Clin Transl Neurol 2014; 1(10): 822-32.
[http://dx.doi.org/10.1002/acn3.120] [PMID: 25493273]
[42]
Brody DL, Holtzman DM. Active and passive immunotherapy for neurodegenerative disorders. Annu Rev Neurosci 2008; 31(1): 175-93.
[http://dx.doi.org/10.1146/annurev.neuro.31.060407.125529] [PMID: 18352830]
[43]
Söderberg L, Johannesson M, Nygren P, et al. Lecanemab, aducanumab, and gantenerumab - binding profiles to different forms of amyloid-beta might explain efficacy and side effects in clinical trials for Alzheimer’s Disease. Neurotherapeutics 2022; 20: 195-206.
[http://dx.doi.org/10.1007/s13311-022-01308-6] [PMID: 36253511]
[44]
van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in Early Alzheimer’s Disease. N Engl J Med 2023; 388(1): 9-21.
[http://dx.doi.org/10.1056/NEJMoa2212948] [PMID: 36449413]
[45]
Lee EY, Srinivasan Y, de Anda J, Nicastro LK, Tükel Ç, Wong GCL. Functional reciprocity of amyloids and antimicrobial peptides: Rethinking the role of supramolecular assembly in host defense, immune activation, and inflammation. Front Immunol 2020; 11: 1629.
[http://dx.doi.org/10.3389/fimmu.2020.01629] [PMID: 32849553]
[46]
Ono K, Tsuji M. Protofibrils of amyloid-β are important targets of a disease-modifying approach for Alzheimer’s Disease. Int J Mol Sci 2020; 21(3): 952.
[http://dx.doi.org/10.3390/ijms21030952] [PMID: 32023927]
[47]
Kile S, Au W, Parise C, et al. IVIG treatment of mild cognitive impairment due to Alzheimer’s disease: A randomised double-blinded exploratory study of the effect on brain atrophy, cognition and conversion to dementia. J Neurol Neurosurg Psychiatry 2017; 88(2): 106-12.
[http://dx.doi.org/10.1136/jnnp-2015-311486] [PMID: 26420886]
[48]
Relkin NR, Thomas RG, Rissman RA, et al. A phase 3 trial of IV immunoglobulin for Alzheimer disease. Neurology 2017; 88(18): 1768-75.
[http://dx.doi.org/10.1212/WNL.0000000000003904] [PMID: 28381506]
[49]
Ahmed MM, Johnson NR, Boyd TD, Coughlan C, Chial HJ, Potter H. Innate immune system activation and neuroinflammation in down syndrome and neurodegeneration: Therapeutic targets or partners? Front Aging Neurosci 2021; 13: 718426.
[http://dx.doi.org/10.3389/fnagi.2021.718426] [PMID: 34603007]
[50]
Potter H, Woodcock JH, Boyd TD, et al. Safety and efficacy of sargramostim (GM‐CSF) in the treatment of Alzheimer’s disease. Alzheimers Dement 2021; 7(1): e12158.
[http://dx.doi.org/10.1002/trc2.12158] [PMID: 33778150]
[51]
Bhattacharya P, Budnick I, Singh M, et al. Dual Role of GM-CSF as a pro-inflammatory and a regulatory cytokine: Implications for immune therapy. J Interferon Cytokine Res 2015; 35(8): 585-99.
[http://dx.doi.org/10.1089/jir.2014.0149] [PMID: 25803788]
[52]
Borriello F, Galdiero M, Varricchi G, Loffredo S, Spadaro G, Marone G. Innate immune modulation by GM-CSF and IL-3 in health and disease. Int J Mol Sci 2019; 20(4): 834.
[http://dx.doi.org/10.3390/ijms20040834] [PMID: 30769926]
[53]
Ahmed MM, Wang ACJ, Elos M, et al. The innate immune system stimulating cytokine GM-CSF improves learning/memory and interneuron and astrocyte brain pathology in Dp16 Down syndrome mice and improves learning/memory in wild-type mice. Neurobiol Dis 2022; 168: 105694.
[http://dx.doi.org/10.1016/j.nbd.2022.105694] [PMID: 35307513]
[54]
Meier-Stephenson FS, Meier-Stephenson VC, Carter MD, et al. Alzheimer’s disease as an autoimmune disorder of innate immunity endogenously modulated by tryptophan metabolites. Alzheimers Dement 2022; 8(1): e12283.
[http://dx.doi.org/10.1002/trc2.12283] [PMID: 35415204]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy