Age-dependency of terminal ileum tissue resident memory T cell responsiveness profiles to S. Typhi following oral Ty21a immunization in humans

Jayaum S Booth, Eric Goldberg, Seema A Patil, Robin S Barnes, Bruce D Greenwald, Marcelo B Sztein, Jayaum S Booth, Eric Goldberg, Seema A Patil, Robin S Barnes, Bruce D Greenwald, Marcelo B Sztein

Abstract

Background: The impact of aging on the immune system is unequivocal and results in an altered immune status termed immunosenescence. In humans, the mechanisms of immunosenescence have been examined almost exclusively in blood. However, most immune cells are present in tissue compartments and exhibit differential cell (e.g., memory T cells -TM) subset distributions. Thus, it is crucial to understand immunosenescence in tissues, especially those that are exposed to pathogens (e.g., intestine). Using a human model of oral live attenuated typhoid vaccine, Ty21a, we investigated the effect of aging on terminal ileum (TI) tissue resident memory T (TRM) cells. TRM provide immediate adaptive effector immune responsiveness at the infection site. However, it is unknown whether aging impacts TRM S. Typhi-responsive cells at the site of infection (e.g., TI). Here, we determined the effect of aging on the induction of TI S. Typhi-responsive TRM subsets elicited by Ty21a immunization.

Results: We observed that aging impacts the frequencies of TI-lamina propria mononuclear cells (LPMC) TM and TRM in both Ty21a-vaccinated and control groups. In unvaccinated volunteers, the frequencies of LPMC CD103- CD4+ TRM displayed a positive correlation with age whilst the CD4/CD8 ratio in LPMC displayed a negative correlation with age. We observed that elderly volunteers have weaker S. Typhi-specific mucosal immune responses following Ty21a immunization compared to adults. For example, CD103+ CD4+ TRM showed reduced IL-17A production, while CD103- CD4+ TRM exhibited lower levels of IL-17A and IL-2 in the elderly than in adults following Ty21a immunization. Similar results were observed in LPMC CD8+ TRM and CD103- CD8+ T cell subsets. A comparison of multifunctional (MF) profiles of both CD4+ and CD8+ TRM subsets between elderly and adults also showed significant differences in the quality and quantity of elicited single (S) and MF responses.

Conclusions: Aging influences tissue resident TM S. Typhi-specific responses in the terminal ileum following oral Ty21a-immunization. This study is the first to provide insights in the generation of local vaccine-specific responses in the elderly population and highlights the importance of evaluating tissue immune responses in the context of infection and aging.

Trial registration: This study was approved by the Institutional Review Board and registered on ClinicalTrials.gov (identifier NCT03970304 , Registered 29 May 2019 - Retrospectively registered).

Keywords: Aging; Oral vaccine; Terminal ileum LPMC; Tissue resident memory T cells; Ty21a; Vaccine-induced responses.

Conflict of interest statement

The authors declared no conflict of interest.

Figures

Fig. 1
Fig. 1
Age differences in frequencies of LPMC memory T cells isolated from Ty21a-vaccinated and unvaccinated volunteers. Comparison of TM subsets including TCM (CD62L+ CD45RA−), TEM (CD62L- CD45RA-), TEMRA (CD62L- CD45RA+), and Tnaive (CD62L+ CD45RA+) between adults (A; < 60 yrs.) and elderly (E; ≥60 yrs.) in TI-LPMC (a) CD4+ T and (b) CD8+ T cells obtained from Ty21a-vaccinated (red symbols) and unvaccinated (black symbols) volunteers. Frequencies of (c) TI-LPMC CD4+ CD103+ and CD4+ CD103- TRM and (d) TI-LPMC CD8+ CD69+ CD103+ (CD8+ TRM) and CD8+ CD69+ CD103- T cells were determined and compared between adults and elderly volunteers following Ty21a immunization. Median values for each group are denoted as horizontal black bars. Significant differences are denoted with *P < 0.05, **P < 0.005. Trends to exhibit significance values are indicated by their p-values
Fig. 2
Fig. 2
Age-dependent association of CD4+ and CD8+ TRM frequencies and T cell subsets in terminal ileum mucosa following oral Ty21a immunization. Correlations between age and the frequencies of tissue resident T cells were determined using Pearson’s analysis. a Unvaccinated LPMC CD4+ CD103+ TRM (black symbols), and CD4+ CD103- TRM (blue symbols), as well as Ty21a-vaccinated LPMC CD4+ CD103+ TRM (green symbols) and CD4+ CD103- TRM (red symbols) frequencies were correlated to the age of the volunteers. b Similarly, unvaccinated LPMC CD8+ TRM cells (black symbols), and CD8+ CD103- T cells (blue symbols), and Ty21a-vaccinated LPMC CD8+ TRM (green symbols) and CD8+ CD103- T cells (red symbols) frequencies were correlated to the age of the volunteers. Significant correlations are denoted with * P < 0.05. Trends to exhibit significant correlations are indicated by their p-values
Fig. 3
Fig. 3
Age-dependent correlation of the ratios of CD4 to CD8 cells among CD3+ T cells in peripheral blood and in CD103+ and CD103- subsets in LPMC isolated from TI biopsies. The ratio of CD4 to CD8 cells were correlated to age in (a) PBMC CD3+ T cells, (b) LPMC CD3+ T cells, (c) LPMC CD3+ CD103+ T cells and (d) LPMC CD3+ CD103- T cells. Significant correlations and trends to show correlations are indicated with * and/or their p-value, respectively
Fig. 4
Fig. 4
Multifunctional (MF) and single-expressing (S) S. Typhi-specific CD4+ and CD8+ TEM responses following Ty21a immunization. Net S. Typhi-specific CD4+ and CD8+ TEM responses were calculated using the FCOM function of Winlist and stratified into S and MF responses. Comparison of TI LPMC S. Typhi-specific (a) CD4+ TEM S, (b) CD4+ TEM MF, (c) CD8+ TEM S and (d) CD8+ TEM MF responses between Ty21a-vaccinated (red symbols) and unvaccinated (black symbols) volunteers were determined with significant differences shown (*P < 0.05). Trends to exhibit significance values are indicated by their p-values. Horizontal black bars represent median values
Fig. 5
Fig. 5
Multifunctional (MF) and single-expressing (S) S. Typhi-specific CD4+ TEM responses in adults compared to elderly volunteers following Ty21a immunization. Net S. Typhi-specific CD4+ TEM responses were calculated using the FCOM function of Winlist and stratified into single-expressing (S) and MF responses. Comparison of TI LPMC S. Typhi-specific CD4+ TEM responses between adults (A- < 60 yrs.) and elderly (E- ≥ 60 yrs.) volunteers in (a) CD107a+, (b) INF-γ+, (c) TNF-α+, (d) IL-17A+, (e) IL-2+, and (f) MIP-1β + S and MF responses in Ty21a-vaccinated (red symbols) and unvaccinated (black symbols) volunteers with significant differences shown (*P < 0.05). Trends to exhibit significance values are indicated by their p-values. Horizontal black bars represent median values
Fig. 6
Fig. 6
LPMC multifunctional (MF) and single-expressing (S) S. Typhi–specific CD8+ TEM responses in adults compared to elderly volunteers following Ty21a immunization. Net S. Typhi-specific CD8+ TEM responses were calculated using the FCOM function of Winlist and stratified into S and MF responses. Comparison of TI LPMC S. Typhi-specific CD8+ TEM responses in (a) CD107a+, (b) INF-γ, (c) TNF-α+, (d) IL-17A+, (e) IL-2+, and (f) MIP-1β + S and MF between adults (A- < 60 yrs.) and elderly (E- ≥ 60 yrs.) volunteers that were either orally vaccinated with Ty21a (red symbols) or unvaccinated (black symbols). Trends to exhibit significant values are indicated by their p-values. Horizontal black bars represent median values
Fig. 7
Fig. 7
S. Typhi-specific responses of TI LPMC CD69+ CD103+ CD4+ TRM and CD69+ CD103- CD4+ TRM subsets in healthy adults and elderly volunteers following oral Ty21a-immunization. The net percentages of S. Typhi-specific responses (IFNγ+, IL-17A+, IL-2+, and TNFα+) in (a) CD69+ CD103+ CD4+ TRM and (b) CD69+ CD103- CD4+ TRM subsets were compared between adults (A- < 60 yrs.) and elderly (E- ≥ 60 yrs.) volunteers who were either Ty21a-vaccinated (red symbols) or unvaccinated (black symbols) with significant differences (*P < 0.05) indicated. Trends to exhibit significance are indicated by their p-values. Horizontal bars (black) represent median values
Fig. 8
Fig. 8
S. Typhi-specific responses of terminal ileum LPMC CD8+ TRM and CD8+ CD69+ CD103- T cell subsets in healthy adults and elderly volunteers following oral Ty21a immunization. The net percentages of S. Typhi-specific responses (IFNγ+, IL-17A+, IL-2+, and TNFα+) in (a) CD8+ TRM and (b) CD8+ CD69+ CD103- T cell subsets were compared between adults (A- < 60 yrs) and elderly (E- ≥ 60 yrs.) volunteers who were either Ty21a-vaccinated (red symbols) or unvaccinated (black symbols). Significant differences (*P < 0.05; **P < 0.005; ***P < 0.0005) are indicated. Trends to exhibit significance are indicated by their p-values. Horizontal bars (black) represent median values

References

    1. Aw D, Silva AB, Palmer DB. Immunosenescence: emerging challenges for an ageing population. Immunology. 2007;120(4):435–446. doi: 10.1111/j.1365-2567.2007.02555.x.
    1. Bektas A, Schurman SH, Sen R, Ferrucci L. Human T cell immunosenescence and inflammation in aging. J Leukoc Biol. 2017;102(4):977–988. doi: 10.1189/jlb.3RI0716-335R.
    1. Scholz JL, Diaz A, Riley RL, Cancro MP, Frasca D. A comparative review of aging and B cell function in mice and humans. Curr Opin Immunol. 2013;25(4):504–510. doi: 10.1016/j.coi.2013.07.006.
    1. Nikolich-Zugich J. Aging of the T cell compartment in mice and humans: from no naive expectations to foggy memories. J Immunol. 2014;193(6):2622–2629. doi: 10.4049/jimmunol.1401174.
    1. Gozalo PL, Pop-Vicas A, Feng Z, Gravenstein S, Mor V. Effect of influenza on functional decline. J Am Geriatr Soc. 2012;60(7):1260–1267. doi: 10.1111/j.1532-5415.2012.04048.x.
    1. Johnson RW, Bouhassira D, Kassianos G, Leplege A, Schmader KE, Weinke T. The impact of herpes zoster and post-herpetic neuralgia on quality-of-life. BMC Med. 2010;8(1):37. doi: 10.1186/1741-7015-8-37.
    1. Wroe PC, Finkelstein JA, Ray GT, Linder JA, Johnson KM, Rifas-Shiman S, Moore MR, Huang SS. Aging population and future burden of pneumococcal pneumonia in the United States. J Infect Dis. 2012;205(10):1589–1592. doi: 10.1093/infdis/jis240.
    1. Wagner A, Garner-Spitzer E, Jasinska J, Kollaritsch H, Stiasny K, Kundi M, Wiedermann U. Age-related differences in humoral and cellular immune responses after primary immunisation: indications for stratified vaccination schedules. Sci Rep. 2018;8(1):9825. doi: 10.1038/s41598-018-28111-8.
    1. Tseng HF, Harpaz R, Luo Y, Hales CM, Sy LS, Tartof SY, Bialek S, Hechter RC, Jacobsen SJ. Declining effectiveness of herpes zoster vaccine in adults aged >/=60 years. J Infect Dis. 2016;213(12):1872–1875. doi: 10.1093/infdis/jiw047.
    1. Edelman R, Deming ME, Toapanta FR, Heuser MD, Chrisley L, Barnes RS, Wasserman SS, Blackwelder WC, Handwerger BS, Pasetti M, Siddiqui KM, Sztein MB. The SENIEUR protocol and the efficacy of hepatitis B vaccination in healthy elderly persons by age, gender, and vaccine route. Immun Ageing. 2020;17(1):9. doi: 10.1186/s12979-020-00179-9.
    1. Chen WH, Kozlovsky BF, Effros RB, Grubeck-Loebenstein B, Edelman R, Sztein MB. Vaccination in the elderly: an immunological perspective. Trends Immunol. 2009;30(7):351–359. doi: 10.1016/j.it.2009.05.002.
    1. Yanes RE, Gustafson CE, Weyand CM, Goronzy JJ. Lymphocyte generation and population homeostasis throughout life. Semin Hematol. 2017;54(1):33–38. doi: 10.1053/j.seminhematol.2016.10.003.
    1. Kim C, Fang F, Weyand CM, Goronzy JJ. The life cycle of a T cell after vaccination - where does immune ageing strike? Clin Exp Immunol. 2017;187(1):71–81. doi: 10.1111/cei.12829.
    1. Siegrist CA, Aspinall R. B-cell responses to vaccination at the extremes of age. Nat Rev Immunol. 2009;9(3):185–194. doi: 10.1038/nri2508.
    1. Del Giudice G, Goronzy JJ, Grubeck-Loebenstein B, Lambert PH, Mrkvan T, Stoddard JJ, et al. Fighting against a protean enemy: immunosenescence, vaccines, and healthy aging. NPJ Aging Mech Dis. 2018;4(1):1. doi: 10.1038/s41514-017-0020-0.
    1. Quinn KM, Fox A, Harland KL, Russ BE, Li J, Nguyen THO, Loh L, Olshanksy M, Naeem H, Tsyganov K, Wiede F, Webster R, Blyth C, Sng XYX, Tiganis T, Powell D, Doherty PC, Turner SJ, Kedzierska K, la Gruta NL. Age-related decline in primary CD8(+) T cell responses is associated with the development of senescence in virtual memory CD8(+) T cells. Cell Rep. 2018;23(12):3512–3524. doi: 10.1016/j.celrep.2018.05.057.
    1. Thome JJ, Farber DL. Emerging concepts in tissue-resident T cells: lessons from humans. Trends Immunol. 2015;36(7):428–435. doi: 10.1016/j.it.2015.05.003.
    1. Mueller SN, Mackay LK. Tissue-resident memory T cells: local specialists in immune defence. Nat Rev Immunol. 2016;16(2):79–89. doi: 10.1038/nri.2015.3.
    1. Kumar BV, Ma W, Miron M, Granot T, Guyer RS, Carpenter DJ, Senda T, Sun X, Ho SH, Lerner H, Friedman AL, Shen Y, Farber DL. Human tissue-resident memory T cells are defined by Core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 2017;20(12):2921–2934. doi: 10.1016/j.celrep.2017.08.078.
    1. Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJ, et al. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity. 2013;38(1):187–197. doi: 10.1016/j.immuni.2012.09.020.
    1. Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med. 2015;7(279):279ra39. doi: 10.1126/scitranslmed.3010302.
    1. Hombrink P, Helbig C, Backer RA, Piet B, Oja AE, Stark R, Brasser G, Jongejan A, Jonkers RE, Nota B, Basak O, Clevers HC, Moerland PD, Amsen D, van Lier RAW. Programs for the persistence, vigilance and control of human CD8(+) lung-resident memory T cells. Nat Immunol. 2016;17(12):1467–1478. doi: 10.1038/ni.3589.
    1. Levine MM, Ferreccio C, Abrego P, Martin OS, Ortiz E, Cryz S. Duration of efficacy of Ty21a, attenuated Salmonella typhi live oral vaccine. Vaccine. 1999;17(Suppl 2):S22–S27. doi: 10.1016/S0264-410X(99)00231-5.
    1. Levine MM. Typhoid fever vaccines. In: Plotkin S, Orenstein WA, Offit PA, Edwards KM, editors. Plokin's Vaccines. 7. Philadelphia: Elsevier; 2018. pp. 1114–1144.
    1. Booth JS, Goldberg E, Patil SA, Barnes RS, Greenwald BD, Sztein MB. Effect of live oral attenuated typhoid vaccine, Ty21a, on systemic and terminal ileum mucosal CD4+ T memory responses in humans. Int Immunol. 2018;31(2):101. doi: 10.1093/intimm/dxy070.
    1. Booth JS, Patil SA, Goldberg E, Barnes RS, Greenwald BD, Sztein MB. Attenuated Oral typhoid vaccine Ty21a elicits Lamina Propria and intra-epithelial lymphocyte tissue-resident effector memory CD8 T responses in the human terminal ileum. Front Immunol. 2019;10:424. doi: 10.3389/fimmu.2019.00424.
    1. Booth JS, Goldberg E, Patil SA, Greenwald BD, Sztein MB. Association between S. Typhi-specific memory CD4+ and CD8+ T responses in the terminal ileum mucosa and in peripheral blood elicited by the live oral typhoid vaccine Ty21a in humans. Hum Vaccin Immunother. 2019;15:1–12. doi: 10.1080/21645515.2018.1564570.
    1. Booth JS, Goldberg E, Barnes RS, Greenwald BD, Sztein MB. Oral typhoid vaccine Ty21a elicits antigen-specific resident memory CD4(+) T cells in the human terminal ileum lamina propria and epithelial compartments. J Transl Med. 2020;18(1):102. doi: 10.1186/s12967-020-02263-6.
    1. Nickerson KP, Senger S, Zhang Y, Lima R, Patel S, Ingano L, Flavahan WA, Kumar DKV, Fraser CM, Faherty CS, Sztein MB, Fiorentino M, Fasano A. Salmonella Typhi colonization provokes extensive transcriptional changes aimed at evading host mucosal immune defense during early infection of human intestinal tissue. EBioMedicine. 2018;31:92–109. doi: 10.1016/j.ebiom.2018.04.005.
    1. Kantele A, Hakkinen M, Moldoveanu Z, Lu A, Savilahti E, Alvarez RD, et al. Differences in immune responses induced by oral and rectal immunizations with Salmonella typhi Ty21a: evidence for compartmentalization within the common mucosal immune system in humans. Infect Immun. 1998;66(12):5630–5635. doi: 10.1128/IAI.66.12.5630-5635.1998.
    1. Vasson MP, Farges MC, Goncalves-Mendes N, Talvas J, Ribalta J, Winklhofer-Roob B, Rock E, Rossary A. Does aging affect the immune status? A comparative analysis in 300 healthy volunteers from France. Austria and Spain Immun Ageing. 2013;10(1):38. doi: 10.1186/1742-4933-10-38.
    1. Strindhall J, Skog M, Ernerudh J, Bengner M, Lofgren S, Matussek A, et al. The inverted CD4/CD8 ratio and associated parameters in 66-year-old individuals: the Swedish HEXA immune study. Age (Dordr) 2013;35(3):985–991. doi: 10.1007/s11357-012-9400-3.
    1. Li M, Yao D, Zeng X, Kasakovski D, Zhang Y, Chen S, Zha X, Li Y, Xu L. Age related human T cell subset evolution and senescence. Immun Ageing. 2019;16(1):24. doi: 10.1186/s12979-019-0165-8.
    1. Lin Y, Kim J, Metter EJ, Nguyen H, Truong T, Lustig A, Ferrucci L, Weng NP. Changes in blood lymphocyte numbers with age in vivo and their association with the levels of cytokines/cytokine receptors. Immun Ageing. 2016;13(1):24. doi: 10.1186/s12979-016-0079-7.
    1. Derhovanessian E, Larbi A, Pawelec G. Biomarkers of human immunosenescence: impact of Cytomegalovirus infection. Curr Opin Immunol. 2009;21(4):440–445. doi: 10.1016/j.coi.2009.05.012.
    1. Booth JS, Patil SA, Ghazi L, Barnes R, Fraser CM, Fasano A, Greenwald BD, Sztein MB. Systemic and terminal ileum mucosal immunity elicited by Oral immunization with the Ty21a typhoid vaccine in humans. Cell Mol Gastroenterol Hepatol. 2017;4(3):419–437. doi: 10.1016/j.jcmgh.2017.08.002.
    1. Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, Flynn BJ, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med. 2007;13(7):843–850. doi: 10.1038/nm1592.
    1. Lindenstrom T, Agger EM, Korsholm KS, Darrah PA, Aagaard C, Seder RA, et al. Tuberculosis subunit vaccination provides long-term protective immunity characterized by multifunctional CD4 memory T cells. J Immunol. 2009;182(12):8047–8055. doi: 10.4049/jimmunol.0801592.
    1. Lin L, Finak G, Ushey K, Seshadri C, Hawn TR, Frahm N, Scriba TJ, Mahomed H, Hanekom W, Bart PA, Pantaleo G, Tomaras GD, Rerks-Ngarm S, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Michael NL, Kim JH, Robb ML, O'Connell RJ, Karasavvas N, Gilbert P, C de Rosa S, McElrath MJ, Gottardo R. COMPASS identifies T-cell subsets correlated with clinical outcomes. Nat Biotechnol. 2015;33(6):610–616. doi: 10.1038/nbt.3187.
    1. Fresnay S, McArthur MA, Magder L, Darton TC, Jones C, Waddington CS, et al. Salmonella Typhi-specific multifunctional CD8+ T cells play a dominant role in protection from typhoid fever in humans. J Transl Med. 2016;14(1):62. doi: 10.1186/s12967-016-0819-7.
    1. Fresnay S, McArthur MA, Magder LS, Darton TC, Jones C, Waddington CS, et al. Importance of Salmonella Typhi-responsive CD8+ T cell immunity in a human typhoid fever challenge model. Front Immunol. 2017;8:208. doi: 10.3389/fimmu.2017.00208.
    1. Burel JG, Apte SH, Groves PL, McCarthy JS, Doolan DL. Polyfunctional and IFN-gamma monofunctional human CD4(+) T cell populations are molecularly distinct. JCI Insight. 2017;2(3):e87499. doi: 10.1172/jci.insight.87499.
    1. Oja AE, Piet B, Helbig C, Stark R, van der Zwan D, Blaauwgeers H, Remmerswaal EBM, Amsen D, Jonkers RE, Moerland PD, Nolte MA, van Lier RAW, Hombrink P. Trigger-happy resident memory CD4(+) T cells inhabit the human lungs. Mucosal Immunol. 2018;11(3):654–667. doi: 10.1038/mi.2017.94.
    1. Park CO, Fu X, Jiang X, Pan Y, Teague JE, Collins N, Tian T, O'Malley JT, Emerson RO, Kim JH, Jung Y, Watanabe R, Fuhlbrigge RC, Carbone FR, Gebhardt T, Clark RA, Lin CP, Kupper TS. Staged development of long-lived T-cell receptor alphabeta TH17 resident memory T-cell population to Candida albicans after skin infection. J Allergy Clin Immunol. 2018;142(2):647–662. doi: 10.1016/j.jaci.2017.09.042.
    1. Goronzy JJ, Weyand CM. Successful and maladaptive T cell aging. Immunity. 2017;46(3):364–378. doi: 10.1016/j.immuni.2017.03.010.
    1. Haynes L, Eaton SM, Burns EM, Randall TD, Swain SL. CD4 T cell memory derived from young naive cells functions well into old age, but memory generated from aged naive cells functions poorly. Proc Natl Acad Sci U S A. 2003;100(25):15053–15058. doi: 10.1073/pnas.2433717100.
    1. Haynes L, Maue AC. Effects of aging on T cell function. Curr Opin Immunol. 2009;21(4):414–417. doi: 10.1016/j.coi.2009.05.009.
    1. Effros RB, Cai Z, Linton PJ. CD8 T cells and aging. Crit Rev Immunol. 2003;23(1–2):45–64. doi: 10.1615/CritRevImmunol.v23.i12.30.
    1. Koch S, Solana R, Dela Rosa O, Pawelec G. Human cytomegalovirus infection and T cell immunosenescence: a mini review. Mech Ageing Dev. 2006;127(6):538–543. doi: 10.1016/j.mad.2006.01.011.
    1. Tu W, Rao S. Mechanisms underlying T cell Immunosenescence: aging and Cytomegalovirus infection. Front Microbiol. 2016;7:2111. doi: 10.3389/fmicb.2016.02111.
    1. Muruganandah V, Sathkumara HD, Navarro S, Kupz A. A systematic review: the role of resident memory T cells in infectious diseases and their relevance for vaccine development. Front Immunol. 2018;9:1574. doi: 10.3389/fimmu.2018.01574.
    1. Shin H, Iwasaki A. Tissue-resident memory T cells. Immunol Rev. 2013;255(1):165–181. doi: 10.1111/imr.12087.
    1. Schenkel JM, Masopust D. Tissue-resident memory T cells. Immunity. 2014;41(6):886–897. doi: 10.1016/j.immuni.2014.12.007.
    1. Douek DC, McFarland RD, Keiser PH, Gage EA, Massey JM, Haynes BF, et al. Changes in thymic function with age and during the treatment of HIV infection. Nature. 1998;396(6712):690–695. doi: 10.1038/25374.
    1. Dock J, Ramirez CM, Hultin L, Hausner MA, Hultin P, Elliott J, Yang OO, Anton PA, Jamieson BD, Effros RB. Distinct aging profiles of CD8+ T cells in blood versus gastrointestinal mucosal compartments. PLoS One. 2017;12(8):e0182498. doi: 10.1371/journal.pone.0182498.
    1. Sansoni P, Vescovini R, Fagnoni F, Biasini C, Zanni F, Zanlari L, Telera A, Lucchini G, Passeri G, Monti D, Franceschi C, Passeri M. The immune system in extreme longevity. Exp Gerontol. 2008;43(2):61–65. doi: 10.1016/j.exger.2007.06.008.
    1. van den Berg SPH, Wong A, Hendriks M, Jacobi RHJ, van Baarle D, van Beek J. Negative effect of age, but not of latent Cytomegalovirus infection on the antibody response to a novel influenza vaccine strain in healthy adults. Front Immunol. 2018;9:82. doi: 10.3389/fimmu.2018.00082.
    1. Roukens AH, Soonawala D, Joosten SA, de Visser AW, Jiang X, Dirksen K, de Gruijter M, van Dissel JT, Bredenbeek PJ, Visser LG. Elderly subjects have a delayed antibody response and prolonged viraemia following yellow fever vaccination: a prospective controlled cohort study. PLoS One. 2011;6(12):e27753. doi: 10.1371/journal.pone.0027753.
    1. Djennad A, Ramsay ME, Pebody R, Fry NK, Sheppard C, Ladhani SN, Andrews NJ. Effectiveness of 23-Valent polysaccharide pneumococcal vaccine and changes in invasive pneumococcal disease incidence from 2000 to 2017 in those aged 65 and over in England and Wales. EClinicalMedicine. 2018;6:42–50. doi: 10.1016/j.eclinm.2018.12.007.
    1. Chelimo K, Embury PB, Sumba PO, Vulule J, Ofulla AV, Long C, et al. Age-related differences in naturally acquired T cell memory to plasmodium falciparum merozoite surface protein 1. PLoS One. 2011;6(9):e24852. doi: 10.1371/journal.pone.0024852.
    1. Schulz AR, Malzer JN, Domingo C, Jurchott K, Grutzkau A, Babel N, et al. Low Thymic activity and dendritic cell numbers are associated with the immune response to primary viral infection in elderly humans. J Immunol. 2015;195(10):4699–4711. doi: 10.4049/jimmunol.1500598.
    1. McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, et al. T cell responses are better correlates of vaccine protection in the elderly. J Immunol. 2006;176(10):6333–6339. doi: 10.4049/jimmunol.176.10.6333.
    1. Senda T, Dogra P, Granot T, Furuhashi K, Snyder ME, Carpenter DJ, Szabo PA, Thapa P, Miron M, Farber DL. Microanatomical dissection of human intestinal T-cell immunity reveals site-specific changes in gut-associated lymphoid tissues over life. Mucosal Immunol. 2019;12(2):378–389. doi: 10.1038/s41385-018-0110-8.
    1. Nguyen TH, McAuley JL, Kim Y, Zheng MZ, Gherardin NA, Godfrey DI, Purcell DF, Sullivan LC, Westall GP, Reading PC, Kedzierska K, Wakim LM. Influenza, but not SARS-CoV-2, infection induces a rapid interferon response that wanes with age and diminished tissue-resident memory CD8(+) T cells. Clin Transl Immunology. 2021;10(1):e1242. doi: 10.1002/cti2.1242.
    1. Oh SJ, Lee JK, Shin OS. Aging and the immune system: the impact of Immunosenescence on viral infection. Immunity Vaccine Immunogenicity Immune Netw. 2019;19(6):e37. doi: 10.4110/in.2019.19.e37.
    1. Ciabattini A, Nardini C, Santoro F, Garagnani P, Franceschi C, Medaglini D. Vaccination in the elderly: the challenge of immune changes with aging. Semin Immunol. 2018;40:83–94. doi: 10.1016/j.smim.2018.10.010.
    1. Jackson SE, Sedikides GX, Okecha G, Poole EL, Sinclair JH, Wills MR. Latent Cytomegalovirus (CMV) infection does not detrimentally Alter T cell responses in the healthy old, But Increased Latent CMV Carriage Is Related to Expanded CMV-Specific T Cells. Front Immunol. 2017;8:733. doi: 10.3389/fimmu.2017.00733.
    1. Staras SA, Dollard SC, Radford KW, Flanders WD, Pass RF, Cannon MJ. Seroprevalence of cytomegalovirus infection in the United States, 1988-1994. Clin Infect Dis. 2006;43(9):1143–1151. doi: 10.1086/508173.
    1. Pawelec G, McElhaney JE, Aiello AE, Derhovanessian E. The impact of CMV infection on survival in older humans. Curr Opin Immunol. 2012;24(4):507–511. doi: 10.1016/j.coi.2012.04.002.
    1. Wikby A, Johansson B, Olsson J, Lofgren S, Nilsson BO, Ferguson F. Expansions of peripheral blood CD8 T-lymphocyte subpopulations and an association with cytomegalovirus seropositivity in the elderly: the Swedish NONA immune study. Exp Gerontol. 2002;37(2–3):445–453. doi: 10.1016/S0531-5565(01)00212-1.
    1. Derhovanessian E, Maier AB, Hahnel K, McElhaney JE, Slagboom EP, Pawelec G. Latent infection with cytomegalovirus is associated with poor memory CD4 responses to influenza a core proteins in the elderly. J Immunol. 2014;193(7):3624–3631. doi: 10.4049/jimmunol.1303361.
    1. Ferreccio C, Levine MM, Rodriguez H, Contreras R. Comparative efficacy of two, three, or four doses of TY21a live oral typhoid vaccine in enteric-coated capsules: a field trial in an endemic area. J Infect Dis. 1989;159(4):766–769. doi: 10.1093/infdis/159.4.766.
    1. Sztein MB, Tanner MK, Polotsky Y, Orenstein JM, Levine MM. Cytotoxic T lymphocytes after oral immunization with attenuated vaccine strains of Salmonella typhi in humans. J Immunol. 1995;155(8):3987–3993.
    1. Booth JS, Toapanta FR, Salerno-Goncalves R, Patil S, Kader HA, Safta AM, et al. Characterization and functional properties of gastric tissue-resident memory T cells from children, adults, and the elderly. Front Immunol. 2014;5:294. doi: 10.3389/fimmu.2014.00294.
    1. Booth JS, Salerno-Goncalves R, Blanchard TG, Patil SA, Kader HA, Safta AM, et al. Mucosal-associated invariant T cells in the human gastric mucosa and blood: role in helicobacter pylori infection. Front Immunol. 2015;6:466. doi: 10.3389/fimmu.2015.00466.
    1. Eiras P, Roldan E, Camarero C, Olivares F, Bootello A, Roy G. Flow cytometry description of a novel CD3−/CD7+ intraepithelial lymphocyte subset in human duodenal biopsies: potential diagnostic value in coeliac disease. Cytometry. 1998;34(2):95–102. doi: 10.1002/(SICI)1097-0320(19980415)34:2<95::AID-CYTO6>;2-B.
    1. Salerno-Goncalves R, Fernandez-Vina M, Lewinsohn DM, Sztein MB. Identification of a human HLA-E-restricted CD8+ T cell subset in volunteers immunized with Salmonella enterica serovar Typhi strain Ty21a typhoid vaccine. J Immunol. 2004;173(9):5852–5862. doi: 10.4049/jimmunol.173.9.5852.

Source: PubMed

3
Abonnieren