Defective T Memory Cell Differentiation after Varicella Zoster Vaccination in Older Individuals

Qian Qi, Mary M Cavanagh, Sabine Le Saux, Lisa E Wagar, Sally Mackey, Jinyu Hu, Holden Maecker, Gary E Swan, Mark M Davis, Cornelia L Dekker, Lu Tian, Cornelia M Weyand, Jörg J Goronzy, Qian Qi, Mary M Cavanagh, Sabine Le Saux, Lisa E Wagar, Sally Mackey, Jinyu Hu, Holden Maecker, Gary E Swan, Mark M Davis, Cornelia L Dekker, Lu Tian, Cornelia M Weyand, Jörg J Goronzy

Abstract

Vaccination with attenuated live varicella zoster virus (VZV) can prevent zoster reactivation, but protection is incomplete especially in an older population. To decipher the molecular mechanisms underlying variable vaccine responses, T- and B-cell responses to VZV vaccination were examined in individuals of different ages including identical twin pairs. Contrary to the induction of VZV-specific antibodies, antigen-specific T cell responses were significantly influenced by inherited factors. Diminished generation of long-lived memory T cells in older individuals was mainly caused by increased T cell loss after the peak response while the expansion of antigen-specific T cells was not affected by age. Gene expression in activated CD4 T cells at the time of the peak response identified gene modules related to cell cycle regulation and DNA repair that correlated with the contraction phase of the T cell response and consequently the generation of long-lived memory cells. These data identify cell cycle regulatory mechanisms as targets to reduce T cell attrition in a vaccine response and to improve the generation of antigen-specific T cell memory, in particular in an older population.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Influence of pre-existing VZV immunity…
Fig 1. Influence of pre-existing VZV immunity on vaccine responses.
(A) VZV-specific T cell frequencies were determined by IFN-γ–specific ELISpot before (day 0) and at days 8±1, 14±1 and 28±3 after vaccination. **p2 = 0.73, p <0.001), but not with initial VZV-specific T cell frequencies (D, p = 0.16). (E and F) Fold change in VZV-specific T cell frequencies from day 0 to day 28 showed no correlation with initial VZV-specific antibodies (E, p = 0.47) or initial T cell frequencies (F, p = 0.12). (G) Fold change in VZV-specific antibodies from day 0 to day 28 did not correlate with fold change in VZV-specific T cell frequencies from day 0 to day 28 (p = 0.38).
Fig 2. Contribution of initial expansion and…
Fig 2. Contribution of initial expansion and subsequent contraction of VZV-specific T cells after vaccination to final T cell memory frequencies.
VZV-specific T cell frequencies were measured by IFN-γ–specific ELISpot. Peak ELISpot counts were defined as the highest observed count at either day 8 (25/29 people) or day 14 (4/29 people) after vaccination. Absolute increase to the peak value (expansion) was calculated compared to day 0 counts. Absolute decline from peak (contraction) was calculated compared to day 28 counts. (A) Unsupervised hierarchical clustering analysis based on frequencies of VZV-specific T cells at all three time points. Vaccinees separated into three main clusters [C1 (black), C2 (red), C3 (blue)]. The color code for the different clusters is maintained in Fig 2B–2F. (B) Mean±SD of VZV-specific T cell frequencies at day 0, peak, and day 28 are shown for each cluster. (C-F) Boxplots showing overall increases in VZV-specific T cell frequencies from day 0 to day 28 (C), expansion in VZV-specific T cell frequencies from day 0 to peak value (D), contraction in VZV-specific T cell frequencies from peak to day 28 (E), and age range of individuals in each cluster (F). p-values calculated by one-tailed Wilcoxon-Mann-Whitney test are shown. (G) Vaccinees were grouped according to their age 59 (green) years and T cell frequency trajectories after vaccinations are shown as described in Fig 2B. (H) When corrected for age, VZV-specific T cell expansion showed a weak correlation with overall increase in VZV-specific T cell frequency from day 0 to day 28 that did not reach significance (r = 0.31, p = 0.10). (I) Contraction after peak responses corrected for age inversely correlated with overall increase in VZV-specific T cell frequency from day 0 to day 28 (r = -0.53, p = 0.003).
Fig 3. T cell responses to VZV…
Fig 3. T cell responses to VZV vaccination are more similar in identical twins than in non-twins.
Pairwise comparisons of fold changes in VZV-specific T cell frequencies (A, C, D) and antibodies (B) were performed for all individuals. Histograms show the frequency distributions of the differences in fold changes for each pair of individuals. Red lines show the position of identical twin comparisons. (A) T cell responses (day 28 to day 0) were more similar between identical twins than between unrelated individuals (p = 0.008). (B) Antibody responses were no more similar between twins than between unrelated individuals (p = 0.44). (C) Effector cell expansion (day 0 to peak) in twins was more similar compared with non-twins with two notable outliers, therefore not reaching significance (p = 0.28). (D) Contraction (peak to day 28) was slightly more similar between twins than between non-twins without reaching significance (p = 0.16).
Fig 4. Correlation of whole blood-derived gene…
Fig 4. Correlation of whole blood-derived gene signatures with VZV-specific T cell responses.
(A) Deconvolution of whole blood gene expression for leukocyte subsets was performed. Volcano plots show fold change of gene expression in monocytes (left), lymphocytes (middle) and neutrophils (right) between day 0 and day 1. The genes with significant changes in expression (fold change>1.5, p

Fig 5. Lasso cytokine predictors of antigen-specific…

Fig 5. Lasso cytokine predictors of antigen-specific T cell expansion and contraction after VZV vaccination.

Fig 5. Lasso cytokine predictors of antigen-specific T cell expansion and contraction after VZV vaccination.
(A) Principal component analysis of serum concentrations of 51 cytokines on day 0 and day 1 was performed. The box plots show the log transformed fold changes (day 1 to day 0) of PC1 (explaining 60% of variation, left panel) and PC2 (9%, right panel) among individuals. PC2 significantly changes between before and day 1 after vaccination (p = 0.017). (B-E) Panels B and D show the estimated mean square error (y-axis) from a sequence of lasso models in predicting VZV-specific T cell expansion (B) and contraction (D) using the baseline frequencies of VZV-specific T cells and serum cytokine changes between day 0 and day 1 after vaccination. The x-axis represents the log-transformed penalty parameter controlling the model complexity determined by the number of predictors in the model shown on top. Panels C and E plot the predicted vs. the true frequencies after leave-one-out cross-validation for the lasso procedures for VZV-specific T cell expansion (C) and contraction (E). The 45° line is shown for orientation.

Fig 6. Correlation of gene signatures in…

Fig 6. Correlation of gene signatures in activated CD4 T cells with VZV-specific T cell…

Fig 6. Correlation of gene signatures in activated CD4 T cells with VZV-specific T cell responses.
(A) PBMCs of one donor before (day 0) and at days 8 and 14 after vaccination were gated on either total CD4 T cells (grey) or IE63 HLA-DRB1*15:01-tetramer-specific CD4 T cells (red). Expression of CD38 and HLA-DR is shown. (B) Frequencies of CD38+HLA-DR+ cells in total IE63 HLA-DRB1*15:01-tetramer-specific CD4 T cells of three individuals before (day 0) and on days 8 and 14 after vaccination. Time points are compared by paired t-test. (C) CD4+CD38+HLA-DR+ T cells were sorted before (day 0, left panels) and on days 8 (middle panels) and 14 (right panels) after vaccination and arrayed for gene expressions. Expression data were summarized as transcriptional modules and compared to changes in frequencies of VZV-specific T cells determined by IFN-γ ELISpot as shown in Fig 1. Histograms show probability distributions of Pearson correlation coefficients of expressed gene modules (red bars) with decline in VZV-specific T cell frequencies (peak to day 28, upper panels) and with overall increase in frequencies (day 0 to day 28, lower panels). Grey shaded curves represent probability distributions of correlations between random data generated from permutations of module gene labels and sample labels. FDR is calculated using linear regression analysis. The number of significantly associated modules is given as N for p<0.01. (D) The circos plot shows the sharing of significant modules obtained from activated T cells on days 0, 8 and 14. Grey bars show the log2 p-value of correlation between modules and T cell responses. Red and blue circles represent thresholds for modules with significant negative correlation and positive correlation to T cell responses, respectively (p<0.01). Curved lines in the center connect significant modules that are shared. (E) Heat maps show hierarchical clustering of correlation coefficients from the comparisons of gene modules with the fold decrease of peak to day 28 frequencies (A) and the fold increase of day 28 to day 0 frequencies (B). Directionality of correlation coefficients for peak to day 28 contraction was inversed to have equal biological directionality.

Fig 7. Preferential association of cell cycle…

Fig 7. Preferential association of cell cycle and DNA repair pathways with VZV-specific T cell…

Fig 7. Preferential association of cell cycle and DNA repair pathways with VZV-specific T cell responses.
(A) Correlation coefficients of modules that were significantly correlated with the decline in frequencies after peak responses as well as the overall increase from day 0 to day 28 as described in Fig 6, are shown as heat maps. Module nomenclature is from [13]. (B) Networks of genes in modules M22.0 (top left panel), M103 (top right panel) and M4.1 (bottom panel). The sizes of nodes represent the absolute value of correlation coefficients between gene expression level at day 14 after vaccination and peak to day 28 T cell responses. Orange and blue colors indicate positive and negative correlation, respectively. The grey line represents the co-expression relationship as described previously [13]. Gene names for the top genes with the highest correlation coefficient are listed.
All figures (7)
Fig 5. Lasso cytokine predictors of antigen-specific…
Fig 5. Lasso cytokine predictors of antigen-specific T cell expansion and contraction after VZV vaccination.
(A) Principal component analysis of serum concentrations of 51 cytokines on day 0 and day 1 was performed. The box plots show the log transformed fold changes (day 1 to day 0) of PC1 (explaining 60% of variation, left panel) and PC2 (9%, right panel) among individuals. PC2 significantly changes between before and day 1 after vaccination (p = 0.017). (B-E) Panels B and D show the estimated mean square error (y-axis) from a sequence of lasso models in predicting VZV-specific T cell expansion (B) and contraction (D) using the baseline frequencies of VZV-specific T cells and serum cytokine changes between day 0 and day 1 after vaccination. The x-axis represents the log-transformed penalty parameter controlling the model complexity determined by the number of predictors in the model shown on top. Panels C and E plot the predicted vs. the true frequencies after leave-one-out cross-validation for the lasso procedures for VZV-specific T cell expansion (C) and contraction (E). The 45° line is shown for orientation.
Fig 6. Correlation of gene signatures in…
Fig 6. Correlation of gene signatures in activated CD4 T cells with VZV-specific T cell responses.
(A) PBMCs of one donor before (day 0) and at days 8 and 14 after vaccination were gated on either total CD4 T cells (grey) or IE63 HLA-DRB1*15:01-tetramer-specific CD4 T cells (red). Expression of CD38 and HLA-DR is shown. (B) Frequencies of CD38+HLA-DR+ cells in total IE63 HLA-DRB1*15:01-tetramer-specific CD4 T cells of three individuals before (day 0) and on days 8 and 14 after vaccination. Time points are compared by paired t-test. (C) CD4+CD38+HLA-DR+ T cells were sorted before (day 0, left panels) and on days 8 (middle panels) and 14 (right panels) after vaccination and arrayed for gene expressions. Expression data were summarized as transcriptional modules and compared to changes in frequencies of VZV-specific T cells determined by IFN-γ ELISpot as shown in Fig 1. Histograms show probability distributions of Pearson correlation coefficients of expressed gene modules (red bars) with decline in VZV-specific T cell frequencies (peak to day 28, upper panels) and with overall increase in frequencies (day 0 to day 28, lower panels). Grey shaded curves represent probability distributions of correlations between random data generated from permutations of module gene labels and sample labels. FDR is calculated using linear regression analysis. The number of significantly associated modules is given as N for p<0.01. (D) The circos plot shows the sharing of significant modules obtained from activated T cells on days 0, 8 and 14. Grey bars show the log2 p-value of correlation between modules and T cell responses. Red and blue circles represent thresholds for modules with significant negative correlation and positive correlation to T cell responses, respectively (p<0.01). Curved lines in the center connect significant modules that are shared. (E) Heat maps show hierarchical clustering of correlation coefficients from the comparisons of gene modules with the fold decrease of peak to day 28 frequencies (A) and the fold increase of day 28 to day 0 frequencies (B). Directionality of correlation coefficients for peak to day 28 contraction was inversed to have equal biological directionality.
Fig 7. Preferential association of cell cycle…
Fig 7. Preferential association of cell cycle and DNA repair pathways with VZV-specific T cell responses.
(A) Correlation coefficients of modules that were significantly correlated with the decline in frequencies after peak responses as well as the overall increase from day 0 to day 28 as described in Fig 6, are shown as heat maps. Module nomenclature is from [13]. (B) Networks of genes in modules M22.0 (top left panel), M103 (top right panel) and M4.1 (bottom panel). The sizes of nodes represent the absolute value of correlation coefficients between gene expression level at day 14 after vaccination and peak to day 28 T cell responses. Orange and blue colors indicate positive and negative correlation, respectively. The grey line represents the co-expression relationship as described previously [13]. Gene names for the top genes with the highest correlation coefficient are listed.

References

    1. Donahue JG, Choo PW, Manson JE, Platt R. The incidence of herpes zoster. Arch Intern Med. 1995;155(15):1605–9. 10.1001/archinte.1995.00430150071008
    1. Yawn BP, Saddier P, Wollan PC, St Sauver JL, Kurland MJ, Sy LS. A population-based study of the incidence and complication rates of herpes zoster before zoster vaccine introduction. Mayo Clin Proc. 2007;82(11):1341–9. 10.4065/82.11.1341
    1. Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med. 2005;352(22):2271–84. 10.1056/NEJMoa051016
    1. Tseng HF, Smith N, Harpaz R, Bialek SR, Sy LS, Jacobsen SJ. Herpes zoster vaccine in older adults and the risk of subsequent herpes zoster disease. JAMA. 2011;305(2):160–6. 10.1001/jama.2010.1983
    1. Langan SM, Smeeth L, Margolis DJ, Thomas SL. Herpes zoster vaccine effectiveness against incident herpes zoster and post-herpetic neuralgia in an older US population: a cohort study. PLoS Med. 2013;10(4):e1001420 PubMed Central PMCID: PMC3621740. 10.1371/journal.pmed.1001420
    1. Duraisingham SS, Rouphael N, Cavanagh MM, Nakaya HI, Goronzy JJ, Pulendran B. Systems biology of vaccination in the elderly. Curr Top Microbiol Immunol. 2013;363:117–42. 10.1007/82_2012_250
    1. Tsang JS, Schwartzberg PL, Kotliarov Y, Biancotto A, Xie Z, Germain RN, et al. Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell. 2014;157(2):499–513. PubMed Central PMCID: PMC4139290. 10.1016/j.cell.2014.03.031
    1. Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12(8):786–95. PubMed Central PMCID: PMC3140559. 10.1038/ni.2067
    1. Tan Y, Tamayo P, Nakaya H, Pulendran B, Mesirov JP, Haining WN. Gene signatures related to B-cell proliferation predict influenza vaccine-induced antibody response. Eur J Immunol. 2014;44(1):285–95. PubMed Central PMCID: PMC3973429. 10.1002/eji.201343657
    1. Franco LM, Bucasas KL, Wells JM, Nino D, Wang X, Zapata GE, et al. Integrative genomic analysis of the human immune response to influenza vaccination. Elife. 2013;2:e00299 PubMed Central PMCID: PMC3713456. 10.7554/eLife.00299
    1. Furman D, Jojic V, Kidd B, Shen-Orr S, Price J, Jarrell J, et al. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol Syst Biol. 2013;9:659 PubMed Central PMCID: PMC3658270. 10.1038/msb.2013.15
    1. Obermoser G, Presnell S, Domico K, Xu H, Wang Y, Anguiano E, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38(4):831–44. PubMed Central PMCID: PMC3681204. 10.1016/j.immuni.2012.12.008
    1. Li S, Rouphael N, Duraisingham S, Romero-Steiner S, Presnell S, Davis C, et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat Immunol. 2014;15(2):195–204. PubMed Central PMCID: PMC3946932. 10.1038/ni.2789
    1. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009;10(1):116–25. PubMed Central PMCID: PMC4049462. 10.1038/ni.1688
    1. Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, et al. Systems Analysis of Immunity to Influenza Vaccination across Multiple Years and in Diverse Populations Reveals Shared Molecular Signatures. Immunity. 2015;43(6):1186–98. 10.1016/j.immuni.2015.11.012
    1. Thakar J, Mohanty S, West AP, Joshi SR, Ueda I, Wilson J, et al. Aging-dependent alterations in gene expression and a mitochondrial signature of responsiveness to human influenza vaccination. Aging. 2015;7(1):38–52. PubMed Central PMCID: PMC4356402. 10.18632/aging.100720
    1. Levin MJ, Smith JG, Kaufhold RM, Barber D, Hayward AR, Chan CY, et al. Decline in varicella-zoster virus (VZV)-specific cell-mediated immunity with increasing age and boosting with a high-dose VZV vaccine. J Infect Dis. 2003;188(9):1336–44. 10.1086/379048
    1. Hata A, Asanuma H, Rinki M, Sharp M, Wong RM, Blume K, et al. Use of an inactivated varicella vaccine in recipients of hematopoietic-cell transplants. N Engl J Med. 2002;347(1):26–34. 10.1056/NEJMoa013441
    1. Levin MJ, Oxman MN, Zhang JH, Johnson GR, Stanley H, Hayward AR, et al. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J Infect Dis. 2008;197(6):825–35. PubMed Central PMCID: PMC4014857. 10.1086/528696
    1. Kaech SM, Wherry EJ. Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. Immunity. 2007;27(3):393–405. PubMed Central PMCID: PMC3431921. 10.1016/j.immuni.2007.08.007
    1. Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol. 2007;25:171–92. 10.1146/annurev.immunol.25.022106.141548
    1. Williams MA, Ravkov EV, Bevan MJ. Rapid culling of the CD4+ T cell repertoire in the transition from effector to memory. Immunity. 2008;28(4):533–45. PubMed Central PMCID: PMC2391296. 10.1016/j.immuni.2008.02.014
    1. van Leeuwen EM, Sprent J, Surh CD. Generation and maintenance of memory CD4(+) T Cells. Curr Opin Immunol. 2009;21(2):167–72. PubMed Central PMCID: PMC2676210. 10.1016/j.coi.2009.02.005
    1. Araki K, Youngblood B, Ahmed R. The role of mTOR in memory CD8 T-cell differentiation. Immunol Rev. 2010;235(1):234–43. PubMed Central PMCID: PMC3760155. 10.1111/j.0105-2896.2010.00898.x
    1. Sasaki S, He XS, Holmes TH, Dekker CL, Kemble GW, Arvin AM, et al. Influence of prior influenza vaccination on antibody and B-cell responses. PLoS One. 2008;3(8):e2975 PubMed Central PMCID: PMC2500171. 10.1371/journal.pone.0002975
    1. Shen-Orr SS, Tibshirani R, Khatri P, Bodian DL, Staedtler F, Perry NM, et al. Cell type-specific gene expression differences in complex tissues. Nat Methods. 2010;7(4):287–9. PubMed Central PMCID: PMC3699332. 10.1038/nmeth.1439
    1. Akondy RS, Monson ND, Miller JD, Edupuganti S, Teuwen D, Wu H, et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J Immunol. 2009;183(12):7919–30. PubMed Central PMCID: PMC3374958. 10.4049/jimmunol.0803903
    1. Bertoli C, Skotheim JM, de Bruin RA. Control of cell cycle transcription during G1 and S phases. Nature reviews Molecular cell biology. 2013;14(8):518–28. PubMed Central PMCID: PMC4569015. 10.1038/nrm3629
    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):6333–9. 10.4049/jimmunol.176.10.6333
    1. Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, et al. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med. 2013;19(10):1305–12. 10.1038/nm.3350
    1. Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med. 2012;18(2):274–80. 10.1038/nm.2612
    1. Ahlers JD, Belyakov IM. Memories that last forever: strategies for optimizing vaccine T-cell memory. Blood. 2010;115(9):1678–89. PubMed Central PMCID: PMC2920202. 10.1182/blood-2009-06-227546
    1. Qi Q, Cavanagh MM, Saux SL, NamKoong H, Kim C, Turgano E, et al. Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination. Science Translational Medicine. 2016;8(332):332ra46 10.1126/scitranslmed.aaf1725
    1. Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8(4):247–58. 10.1038/nri2274
    1. Klein NP, Holmes TH, Sharp MA, Heineman TC, Schleiss MR, Bernstein DI, et al. Variability and gender differences in memory T cell immunity to varicella-zoster virus in healthy adults. Vaccine. 2006;24(33–34):5913–8. 10.1016/j.vaccine.2006.04.060
    1. Tan PL, Jacobson RM, Poland GA, Jacobsen SJ, Pankratz VS. Twin studies of immunogenicity—determining the genetic contribution to vaccine failure. Vaccine. 2001;19(17–19):2434–9. 10.1016/S0264-410X(00)00468-0
    1. Newport MJ, Goetghebuer T, Weiss HA, Whittle H, Siegrist CA, Marchant A, et al. Genetic regulation of immune responses to vaccines in early life. Genes Immun. 2004;5(2):122–9. 10.1038/sj.gene.6364051
    1. Brodin P, Jojic V, Gao T, Bhattacharya S, Angel CJ, Furman D, et al. Variation in the human immune system is largely driven by non-heritable influences. Cell. 2015;160(1–2):37–47. PubMed Central PMCID: PMC4302727. 10.1016/j.cell.2014.12.020
    1. Wang C, Liu Y, Cavanagh MM, Le Saux S, Qi Q, Roskin KM, et al. B-cell repertoire responses to varicella-zoster vaccination in human identical twins. Proc Natl Acad Sci U S A. 2015;112(2):500–5. PubMed Central PMCID: PMC4299233. 10.1073/pnas.1415875112
    1. Son YM, Ahn SM, Kim GR, Moon YS, Kim SH, Park YM, et al. Resistin enhances the expansion of regulatory T cells through modulation of dendritic cells. BMC Immunol. 2010;11:33 PubMed Central PMCID: PMCPMC2914082. 10.1186/1471-2172-11-33
    1. Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, Masopust D, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 2008;28(5):710–22. 10.1016/j.immuni.2008.02.020
    1. Hodes RJ, Hathcock KS, Weng NP. Telomeres in T and B cells. Nat Rev Immunol. 2002;2(9):699–706. 10.1038/nri890
    1. Sharpless NE, DePinho RA. Telomeres, stem cells, senescence, and cancer. The Journal of clinical investigation. 2004;113(2):160–8. PubMed Central PMCID: PMC311439. 10.1172/JCI20761
    1. Goronzy JJ, Weyand CM. Understanding immunosenescence to improve responses to vaccines. Nat Immunol. 2013;14(5):428–36. PubMed Central PMCID: PMC4183346. 10.1038/ni.2588
    1. Kim C, Jay DC, Williams MA. Dynamic functional modulation of CD4+ T cell recall responses is dependent on the inflammatory environment of the secondary stimulus. PLoS Pathog. 2014;10(5):e1004137 PubMed Central PMCID: PMC4031222. 10.1371/journal.ppat.1004137
    1. Lamar DL, Weyand CM, Goronzy JJ. Promoter choice and translational repression determine cell type-specific cell surface density of the inhibitory receptor CD85j expressed on different hematopoietic lineages. Blood. 2010;115(16):3278–86. PubMed Central PMCID: PMCPMC2858479. 10.1182/blood-2009-09-243493
    1. Mjelle R, Hegre SA, Aas PA, Slupphaug G, Drablos F, Saetrom P, et al. Cell cycle regulation of human DNA repair and chromatin remodeling genes. DNA Repair (Amst). 2015;30:53–67. 10.1016/j.dnarep.2015.03.007
    1. Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461(7267):1071–8. PubMed Central PMCID: PMC2906700. 10.1038/nature08467
    1. Fang F, Yu M, Cavanagh MM, Hutter Saunders J, Qi Q, Ye Z, et al. Expression of CD39 on Activated T Cells Impairs their Survival in Older Individuals. Cell reports. 2016;14(5):1218–31. 10.1016/j.celrep.2016.01.002
    1. Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med. 2014;6(268):268ra179 10.1126/scitranslmed.3009892
    1. Krasnow RE, Jack LM, Lessov-Schlaggar CN, Bergen AW, Swan GE. The Twin Research Registry at SRI International. Twin Res Hum Genet. 2013;16(1):463–70. 10.1017/thg.2012.81
    1. Abbas AR, Wolslegel K, Seshasayee D, Modrusan Z, Clark HF. Deconvolution of blood microarray data identifies cellular activation patterns in systemic lupus erythematosus. PloS one. 2009;4(7):e6098 PubMed Central PMCID: PMC2699551. 10.1371/journal.pone.0006098
    1. Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize implements and enhances circular visualization in R. Bioinformatics. 2014;30(19):2811–2. 10.1093/bioinformatics/btu393

Source: PubMed

3
Abonner