Review Special Issues

The immune system through the ages

  • Received: 07 July 2022 Revised: 01 September 2022 Accepted: 07 September 2022 Published: 14 September 2022
  • The components of the immune system develop in utero and like a computer, some components are immediately functional (the innate components) but other components must learn the programs and details necessary to function (antigen adaptive components). Like other systems, including military and municipal, the innate and antigen specific components develop into an immune system that helps maintain and surveil the other body processes and systems for aberrations, provide surveillance and protection of the mucoepithelial borders and protection from microbial invasion. Inability, excesses, or errors in these processes cause disease. Aging of the immune system brings immunosenescence, inflammaging, more errors, and decreased surveillance which increases risk for new infections (e.g. COVID-19, influenza), recurrence of latent infections, cancer and autoimmune and inflammatory diseases. With greater understanding of the surveillance, effector and regulatory deficits upon aging, better therapies can be developed.

    Citation: Ken S. Rosenthal, Jordan B. Baker. The immune system through the ages[J]. AIMS Allergy and Immunology, 2022, 6(3): 170-187. doi: 10.3934/Allergy.2022013

    Related Papers:

  • The components of the immune system develop in utero and like a computer, some components are immediately functional (the innate components) but other components must learn the programs and details necessary to function (antigen adaptive components). Like other systems, including military and municipal, the innate and antigen specific components develop into an immune system that helps maintain and surveil the other body processes and systems for aberrations, provide surveillance and protection of the mucoepithelial borders and protection from microbial invasion. Inability, excesses, or errors in these processes cause disease. Aging of the immune system brings immunosenescence, inflammaging, more errors, and decreased surveillance which increases risk for new infections (e.g. COVID-19, influenza), recurrence of latent infections, cancer and autoimmune and inflammatory diseases. With greater understanding of the surveillance, effector and regulatory deficits upon aging, better therapies can be developed.



    加载中


    [1] Simon AK, Hollander GA, McMichael A (2015) Evolution of the immune system in humans from infancy to old age. P Roy Soc B-Biol Sci 282: 20143085. https://doi.org/10.1098/rspb.2014.3085
    [2] Nikolich-Žugich J (2018) The twilight of immunity: emerging concepts in aging of the immune system. Nat Immunol 19: 10-19. https://doi.org/10.1038/s41590-017-0006-x
    [3] Weiskopf D, Weinberger B, Grubeck-Loebenstein B (2009) The aging of the immune system. Transplant Int 22: 1041-1050. https://doi.org/10.1111/j.1432-2277.2009.00927.x
    [4] Weyand CM, Goronzy JJ (2016) Aging of the immune system. Mechanisms and therapeutic targets. Ann Am Thorac Soc 13: S422-S428. https://doi.org/10.1513/AnnalsATS.201602-095AW
    [5] Vasto S, Caruso C (2004) Immunity & ageing: a new journal looking at ageing from an immunological point of view. Immun Ageing 1: 1-4. https://doi.org/10.1186/1742-4933-1-1
    [6] Shaw AC, Joshi S, Greenwood H, et al. (2010) Aging of the innate immune system. Curr Opin Immunol 22: 507-513. https://doi.org/10.1016/j.coi.2010.05.003
    [7] Hossain Z, Reza AHMM, Qasem WA, et al. (2022) Development of the immune system in the human embryo. Pediatr Res 2022: 1-5. https://doi.org/10.1038/s41390-022-01940-0
    [8] Márquez EJ, Chung CH, Marches R, et al. (2020) Sexual-dimorphism in human immune system aging. Nat Commun 11: 751. https://doi.org/10.1038/s41467-020-14396-9
    [9] Rosenthal KS (2018) Immune monitoring of the body's borders. AIMS Allergy Immunol 2: 148-164. https://doi.org/10.3934/Allergy.2018.3.148
    [10] Rosenthal KS (2017) Dealing with garbage is the immune system's main job. MOJ Immunol 5: 00174. https://doi.org/10.15406/moji.2017.05.00174
    [11] Melville JM, Moss TJM (2013) The immune consequences of preterm birth. Front Neurosci 7: 79. https://doi.org/10.3389/fnins.2013.00079
    [12] Herzenberg LA, Tung JW (2006) B cell lineages: documented at last!. Nat Immunol 7: 225-226. https://doi.org/10.1038/ni0306-225
    [13] Hornef MW, Torow N (2020) ‘Layered immunity’ and the ‘neonatal window of opportunity’—timed succession of non-redundant phases to establish mucosal host–microbial homeostasis after birth. Immunology 159: 15-25. https://doi.org/10.1111/imm.13149
    [14] Filias A, Theodorou GL, Mouzopoulou S, et al. (2011) Phagocytic ability of neutrophils and monocytes in neonates. BMC Pediatr 11: 29. https://doi.org/10.1186/1471-2431-11-29
    [15] Hebel K, Weinert S, Kuropka B, et al. (2014) CD4+ T cells from human neonates and infants are poised spontaneously to run a nonclassical IL-4 program. J Immunol 192: 5160-5170. https://doi.org/10.4049/jimmunol.1302539
    [16] Leeansyah E, Loh L, Nixon DF, et al. (2014) Acquisition of innate-like microbial reactivity in mucosal tissues during human fetal MAIT-cell development. Nat Commun 5: 3143. https://doi.org/10.1038/ncomms4143
    [17] Silva-Santos B, Schamel WW, Fisch P, et al. (2012) γδ T-cell conference 2012: close encounters for the fifth time. Eur J Immunol 42: 3101-3105. https://doi.org/10.1002/eji.201270101
    [18] Gibbons D, Fleming P, Virasami A, et al. (2014) Interleukin-8 (CXCL8) production is a signatory T cell effector function of human newborn infants. Nat Med 20: 1206-1210. https://doi.org/10.1038/nm.3670
    [19] Gibbons DL, Haque SF, Silberzahn T, et al. (2009) Neonates harbor highly active γδ T cells with selective impairments in preterm infants. Eur J Immunol 39: 1794-1806. https://doi.org/10.1002/eji.200939222
    [20] Pou C, Nkulikiyimfura D, Henckel E, et al. (2019) The repertoire of maternal anti-viral antibodies in human newborns. Nat Med 25: 591-596. https://doi.org/10.1038/s41591-019-0392-8
    [21] Sarvas H, Seppälä I, Kurikka S, et al. (1993) Half-life of the maternal IgG1 allotype in infants. J Clin Immunol 13: 145-151. https://doi.org/10.1007/BF00919271
    [22] Fouda GG, Martinez DR, Swamy GK, et al. (2018) The Impact of IgG transplacental transfer on early life immunity. Immunohorizons 2: 14-25. https://doi.org/10.4049/immunohorizons.1700057
    [23] Katherine Z, Sanidad KZ, Amir M, et al. (2022) Maternal gut microbiome-induced IgG regulates neonatal gut microbiome and immunity. Sci Immunol 7: eabh3816. https://doi.org/10.1126/sciimmunol.abh3816
    [24] Kalliomäki M, Ouwehand A, Arvilommi H, et al. (1999) Transforming growth factor-β in breast milk: a potential regulator of atopic disease at an early age. J Allergy Clin Immunol 104: 1251-1257. https://doi.org/10.1016/S0091-6749(99)70021-7
    [25] Brodin P (2022) Immune-microbe interactions early in life: A determinant of health and disease long term. Science 376: 945-950. https://doi.org/10.1126/science.abk2189
    [26] Olin A, Henckel E, Chen Y, et al. (2018) Stereotypic immune system development in newborn children. Cell 174: 1277-1292. https://doi.org/10.1016/j.cell.2018.06.045
    [27] Johnson JL, Jones MB, Cobb BA (2015) Polysaccharide A from the capsule of Bacteroides fragilis induces clonal CD4+ T cell expansion. J Biol Chem 290: 5007-5014. https://doi.org/10.1074/jbc.M114.621771
    [28] Kelchtermans H, Billiau A, Matthys P (2008) How interferon-γ keeps autoimmune diseases in check. Trends Immunol 29: 479-486. https://doi.org/10.1016/j.it.2008.07.002
    [29] Stiemsma LT, Reynolds LA, Turvey SE, et al. (2015) The hygiene hypothesis: current perspectives and future therapies. Immunotargets Ther 4: 143-157. https://doi.org/10.2147/ITT.S61528
    [30] Okada H, Kuhn C, Feillet H, et al. (2010) The ‘hygiene hypothesis’ for autoimmune and allergic diseases: an update. Clin Exp Immunol 160: 1-9. https://doi.org/10.1111/j.1365-2249.2010.04139.x
    [31] Hill DA, Spergel JM (2018) The atopic march: Critical evidence and clinical relevance. Ann Allerg Asthma Im 120: 131-137. https://doi.org/10.1016/j.anai.2017.10.037
    [32] Neu N, Duchon J, Zachariah P (2015) TORCH Infections. Clin Perinatol 42: 77-103. https://doi.org/10.1016/j.clp.2014.11.001
    [33] Menzies D (2000) What does tuberculin reactivity after bacille Calmette–Guérin vaccination tell us?. Clin Infect Dis 31: S71-S74. https://doi.org/10.1086/314075
    [34] Sherwood ER, Burelbach KR, McBride MA, et al. (2022) Innate immune memory and the host response to infection. J Immunol 208: 785-792. https://doi.org/10.4049/jimmunol.2101058
    [35] Loske J, Röhmel J, Lukassen S, et al. (2022) Pre-activated antiviral innate immunity in the upper airways controls early SARS-CoV-2 infection in children. Nat Biotechnol 40: 319-324. https://doi.org/10.1038/s41587-021-01037-9
    [36] Azzi T, Lünemann A, Murer A, et al. (2014) Role for early-differentiated natural killer cells in infectious mononucleosis. Blood 124: 2533-2543. https://doi.org/10.1182/blood-2014-01-553024
    [37] Takahashi T, Iwasaki A (2021) Sex differences in immune responses. Science 371: 347-348. https://doi.org/10.1126/science.abe7199
    [38] Klein SL, Flanagan KL (2016) Sex differences in immune responses. Nat Rev Immunol 16: 626-638. https://doi.org/10.1038/nri.2016.90
    [39] Klein SL, Jedlicka A, Pekosz A (2010) The Xs and Y of immune responses to viral vaccines. Lancet Infect Dis 10: 338-349. https://doi.org/10.1016/S1473-3099(10)70049-9
    [40] Pinheiro I, Dejager L, Libert C (2011) X-chromosome-located microRNAs in immunity: Might they explain male/female differences?. Bioessays 33: 791-802. https://doi.org/10.1002/bies.201100047
    [41] Griesbeck M, Ziegler S, Laffont S, et al. (2015) Sex differences in plasmacytoid dendritic cell levels of IRF5 drive higher IFN-α production in women. J Immunol 195: 5327-5336. https://doi.org/10.4049/jimmunol.1501684
    [42] Haitao T, Vermunt JV, Abeykoon J, et al. (2020) COVID-19 and sex differences: mechanisms and biomarkers. Mayo Clin Proc 95: 2189-2203. https://doi.org/10.1016/j.mayocp.2020.07.024
    [43] Asano T, Boisson B, Onodi F, et al. (2021) X-linked recessive TLR7 deficiency in ~1% of men under 60 years old with life-threatening COVID-19. Sci Immunol 6: eabl4348. https://doi.org/10.1126/sciimmunol.abl4348
    [44] Aomatsu M, Kato T, Kasahara E, et al. (2013) Gender difference in tumor necrosis factor-α production in human neutrophils stimulated by lipopolysaccharide and interferon-γ. Biochem Bioph Res Co 441: 220-225. https://doi.org/10.1016/j.bbrc.2013.10.042
    [45] Brunsing RL, Owens KS, Prossnitz ER (2013) The G protein-coupled estrogen receptor (GPER) agonist G-1 expands the regulatory T-cell population under TH17-polarizing conditions. J Immunother 36: 190-196. https://doi.org/10.1097/CJI.0b013e31828d8e3b
    [46] Jaillon S, Berthenet K, Garlanda C (2019) Sexual dimorphism in innate immunity. Clin Rev Allerg Immu 56: 308-321. https://doi.org/10.1007/s12016-017-8648-x
    [47] Arruvito L, Sanz M, Banham AH, et al. (2007) Expansion of CD4+CD25+ and FOXP3+ regulatory T cells during the follicular phase of the menstrual cycle: implications for human reproduction. J Immunol 178: 2572-2578. https://doi.org/10.4049/jimmunol.178.4.2572
    [48] Mor G, Aldo P, Alvero A (2017) The unique immunological and microbial aspects of pregnancy. Nat Rev Immunol 17: 469-482. https://doi.org/10.1038/nri.2017.64
    [49] Krop J, Heidt S, Claas FHJ, et al. (2020) Regulatory T cells in pregnancy: it is not all about FoxP3. Front Immunol 11: 1182. https://doi.org/10.3389/fimmu.2020.01182
    [50] Hanna J, Goldman-Wohl D, Hamani Y, et al. (2006) Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat Med 12: 1065-1074. https://doi.org/10.1038/nm1452
    [51] Le Bouteiller P (2013) Human decidual NK cells: unique and tightly regulated effector functions in healthy and pathogen-infected pregnancies. Front Immunol 4: 404. https://doi.org/10.3389/fimmu.2013.00404
    [52] Sojka DK, Yang L, Yokoyama WM (2019) Uterine natural killer cells. Front Immunol 10: 960. https://doi.org/10.3389/fimmu.2019.00960
    [53] Yang SL, Wang HY, Li DJ, et al. (2019) Role of decidual natural killer cells at the maternal–fetal interface during pregnancy. Reprod Dev Med 3: 165-169. https://doi.org/10.4103/2096-2924.268161
    [54] Piccinni MP, Lombardelli L, Logiodice F, et al. (2016) How pregnancy can affect autoimmune diseases progression?. Clin Mol Allergy 14: 11. https://doi.org/10.1186/s12948-016-0048-x
    [55] Bupp MRG, Jorgensen TN (2018) Androgen-induced immunosuppression. Front Immunol 9: 794. https://doi.org/10.3389/fimmu.2018.00794
    [56] Trigunaite A, Dimo J, Jørgensen TN (2015) Suppressive effects of androgens on the immune system. Cellular Immunol 294: 87-94. https://doi.org/10.1016/j.cellimm.2015.02.004
    [57] Angele MK, Pratschke S, Hubbard WJ, et al. (2014) Gender differences in sepsis. Virulence 5: 12-19. https://doi.org/10.4161/viru.26982
    [58] Meier A, Chang J, Chan E, et al. (2009) Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat Med 15: 955-959. https://doi.org/10.1038/nm.2004
    [59] Meester I, Manilla-Muñoz E, León-Cachón, RBR, et al. (2020) SeXY chromosomes and the immune system: reflections after a comparative study. Biol Sex Differ 11: 1-13. https://doi.org/10.1186/s13293-019-0278-y
    [60] Russi AE, Walker-Caulfield ME, Ebel ME, et al. (2015) Cutting edge: c-Kit signaling differentially regulates type 2 innate lymphoid cell accumulation and susceptibility to central nervous system demyelination in male and female SJL mice. J Immunol 194: 5609-5613. https://doi.org/10.4049/jimmunol.1500068
    [61] Zhang MA, Rego D, Moshkova M, et al. (2012) Peroxisome proliferator-activated receptor (PPAR)α and -γ regulate IFNγ and IL-17A production by human T cells in a sex-specific way. P Natl Acad Sci USA 109: 9505-9510. https://doi.org/10.1073/pnas.1118458109
    [62] Tadount F, Doyon-Plourde P, Rafferty E, et al. (2020) Is there a difference in the immune response, efficacy, effectiveness and safety of seasonal influenza vaccine in males and females?—A systematic review. Vaccine 38: 444-459. https://doi.org/10.1016/j.vaccine.2019.10.091
    [63] Gee J, Marquez P, Su J, et al. (2021) First month of COVID-19 vaccine safety monitoring—United States, December 14, 2020-January 13. MMWR Morb Mortal Wkly Rep 70: 283-288. https://doi.org/10.15585/mmwr.mm7008e3
    [64] López-Otín C, Blasco MA, Partridge L, et al. (2013) The hallmarks of aging. Cell 153: 1194-1217. https://doi.org/10.1016/j.cell.2013.05.039
    [65] Cuervo AM, Macian F (2014) Autophagy and the immune function in aging. Curr Opin Immunol 29: 97-104. https://doi.org/10.1016/j.coi.2014.05.006
    [66] Gustafson CE, Kim C, Weyand CM, et al. (2020) Influence of immune aging on vaccine responses. J Allergy Clin Immunol 145: 1309-1321. https://doi.org/10.1016/j.jaci.2020.03.017
    [67] Ciabattini A, Nardini C, Santoro F, et al. (2018) Vaccination in the elderly: The challenge of immune changes with aging. Semin Immunol 40: 83-94. https://doi.org/10.1016/j.smim.2018.10.010
    [68] Fülöp T, Foris G, Worum I, et al. (1985) Age-dependent alterations of Fcγ receptor-mediated effector functions of human polymorphonuclear leucocytes. Clin Exp Immunol 61: 425-432.
    [69] Butcher S, Chahel H, Lord JM (2000) Ageing and the neutrophil: No appetite for killing?. Immunology 100: 411-416. https://doi.org/10.1046/j.1365-2567.2000.00079.x
    [70] Solana R, Tarazona R, Gayoso I, et al. (2012) Innate immunosenescence: Effect of aging on cells and receptors of the innate immune system in humans. Semin Immunol 24: 331-341. https://doi.org/10.1016/j.smim.2012.04.008
    [71] Mylotte JM, Naughton B, Saludades C, et al. (1998) Validation and application of the pneumonia prognosis index to nursing home residents with pneumonia. J Am Geriatr Soc 46: 1538-1544. https://doi.org/10.1111/j.1532-5415.1998.tb01539.x
    [72] Hasegawa T, Feng Z, Yan Z, et al. (2020) Reduction in human epidermal Langerhans cells with age is associated with decline in CXCL14-mediated recruitment of CD14+ monocytes. J Invest Dermatol 140: 1327-1334. https://doi.org/10.1016/j.jid.2019.11.017
    [73] Zegarska B, Pietkun K, Giemza-Kucharska P, et al. (2017) Changes of Langerhans cells during skin ageing. Postepy Dermatol Alergol 34: 260-267. https://doi.org/10.5114/ada.2017.67849
    [74] Salimi S, Hamlyn JM (2020) COVID-19 and crosstalk with the hallmarks of aging. J Gerontol A-Biol 75: e34-e41. https://doi.org/10.1093/gerona/glaa149
    [75] Becklund B, Purton J, Ramsey C, et al. (2016) The aged lymphoid tissue environment fails to support naïve T cell homeostasis. Sci Rep 6: 30842. https://doi.org/10.1038/srep30842
    [76] Thompson HL, Smithey MJ, Surh CD, et al. (2017) Functional and homeostatic impact of age-related changes in lymph node stroma. Front Immunol 8: 706. https://doi.org/10.3389/fimmu.2017.00706
    [77] Naylor K, Li G, Vallejo AN, et al. (2005) The influence of age on T cell generation and TCR diversity. J Immunol 174: 7446-7452. https://doi.org/10.4049/jimmunol.174.11.7446
    [78] Goronzy JJ, Lee WW, Weyand CM (2007) Aging and T-cell diversity. Exp Gerontol 42: 400-406. https://doi.org/10.1016/j.exger.2006.11.016
    [79] Moro-García MA, Alonso-Arias R, López-Vázquez A, et al. (2012) Relationship between functional ability in older people, immune system status, and intensity of response to CMV. Age 34: 479-495. https://doi.org/10.1007/s11357-011-9240-6
    [80] Pangrazzi L, Weinberger BZ (2020) T cells, aging and senescence. Exp Gerontol 134: 110887. https://doi.org/10.1016/j.exger.2020.110887
    [81] Solana R, Tarazona R, Aiello AE, et al. (2012) CMV and Immunosenescence: from basics to clinics. Immun Ageing 9: 23. https://doi.org/10.1186/1742-4933-9-23
    [82] Frasca D, Blomberg BB, Fuldner R, et al. (2018) “Aging and immunity” symposium: Meeting report. Exp Gerontol 105: 1-3. https://doi.org/10.1016/j.exger.2017.12.004
    [83] Goronzy J, Weyand C (2013) Understanding immunosenescence to improve responses to vaccines. Nat Immunol 14: 428-436. https://doi.org/10.1038/ni.2588
    [84] Feehan J, Tripodi N, Apostolopoulos V (2021) The twilight of the immune system: The impact of immunosenescence in aging. Maturitas 147: 7-13. https://doi.org/10.1016/j.maturitas.2021.02.006
    [85] Raynor J, Lages CS, Shehata H, et al. (2012) Homeostasis and function of regulatory T cells in aging. Curr Opin Immunol 24: 482-487. https://doi.org/10.1016/j.coi.2012.04.005
    [86] Neiman M, Hellström C, Just D, et al. (2019) Individual and stable autoantibody repertoires in healthy individuals. Autoimmunity 52: 1-11. https://doi.org/10.1080/08916934.2019.1581774
    [87] Khan MWA, Al Otaibi A, Sherwani S, et al. (2020) Glycation and oxidative stress increase autoantibodies in the elderly. Molecules 25: 3675. https://doi.org/10.3390/molecules25163675
    [88] Barcenas-Morales G, Cortes-Acevedo P, Doffinger R (2019) Anticytokine autoantibodies leading to infection: early recognition, diagnosis and treatment options. Curr Opin Infect Dis 32: 330-336. https://doi.org/10.1097/QCO.0000000000000561
    [89] Merkel PA, Lebo T, Knight V (2019) Functional analysis of anti-cytokine autoantibodies using flow cytometry. Front Immunol 10: 1517. https://doi.org/10.3389/fimmu.2019.01517
    [90] Bastard P, Gervais A, Le Voyeret T, et al. (2021) Autoantibodies neutralizing type I IFNs are present in ~4% of uninfected individuals over 70 years old and account for ~20% of COVID-19 deaths. Sci Immunol 6: eabl4340. https://doi.org/10.1126/sciimmunol.abl4340
    [91] Franceschi C, Salvioli S, Garagnani P, et al. (2017) Immunobiography and the heterogeneity of immune responses in the elderly: a focus on inflammaging and trained immunity. Front Immunol 8: 982. https://doi.org/10.3389/fimmu.2017.00982
    [92] Fulop T, Larbi A, Pawelec G, et al. (2021) Immunology of aging: the birth of inflammaging. Clin Rev Allerg Immu 2021: 1-14. https://doi.org/10.1007/s12016-021-08899-6
    [93] Cai D, Liu T (2012) Inflammatory cause of metabolic syndrome via brain stress and NF-κB. Aging 4: 98-115. https://doi.org/10.18632/aging.100431
    [94] Reed RG (2019) Stress and immunological aging. Curr Opin Behav Sci 28: 38-43. https://doi.org/10.1016/j.cobeha.2019.01.012
    [95] Klopack ET, Crimmins EM, Cole SW, et al. (2022) Social stressors associated with age-related T lymphocyte percentages in older US adults: Evidence from the US Health and Retirement Study. P Natl Acad Sci USA 119: e2202780119. https://doi.org/10.1073/pnas.2202780119
    [96] Gouin JP, Hantsoo L, Kiecolt-Glaser JK (2008) Immune dysregulation and chronic stress among older adults: A review. Neuroimmunomodulat 15: 251-259. https://doi.org/10.1159/000156468
    [97] Minciullo PL, Catalano A, Mandraffino G, et al. (2015) Inflammaging and anti-inflammaging: the role of cytokines in extreme longevity. Arch Immunol Ther Ex 64: 111-126. https://doi.org/10.1007/s00005-015-0377-3
    [98] Tajbakhsh A, Farahani N, Gheibihayat SM, et al. (2021) Autoantigen-specific immune tolerance in pathological and physiological cell death: Nanotechnology comes into view. Int Immunopharmacol 90: 107177. https://doi.org/10.1016/j.intimp.2020.107177
    [99] Giefing-Kröll C, Berger P, Lepperdinger G, et al. (2015) How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging Cell 14: 309-321. https://doi.org/10.1111/acel.12326
    [100] Kauffman CA, Yoshikawa TT (2001) Fungal infections in older adults. Clin Infect Dis 33: 550-555. https://doi.org/10.1086/322685
    [101] Shea KM, Kammerer JS, Winston CA, et al. (2014) Estimated rate of reactivation of latent tuberculosis infection in the United States, overall and by population subgroup. Am J Epidemiol 179: 216-225. https://doi.org/10.1093/aje/kwt246
    [102] Nikolich-Zugich J, Knox KS, Rios CT, et al. (2020) SARS-CoV-2 and COVID-19 in older adults: what we may expect regarding pathogenesis, immune responses, and outcomes. Geroscience 42: 505-514. https://doi.org/10.1007/s11357-020-00186-0
    [103] Boraschi D, Italiani P (2014) Immunosenescence and vaccine failure in the elderly: Strategies for improving response. Immunol Lett 162: 346-353. https://doi.org/10.1016/j.imlet.2014.06.006
    [104] Kogut I, Scholz JL, Cancro MP, et al. (2012) B cell maintenance and function in aging. Semin Immunol 24: 342-349. https://doi.org/10.1016/j.smim.2012.04.004
    [105] Franceschi C, Capri M, Monti D, et al. (2007) Inflammaging and anti-inflammaging: A systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev 128: 92-105. https://doi.org/10.1016/j.mad.2006.11.016
  • Reader Comments
  • © 2022 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1189) PDF downloads(119) Cited by(0)

Article outline

Figures and Tables

Tables(2)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog