Abstract
Influenza vaccination is less effective in elderly as compared to young individuals. Several studies have addressed the identification of immune biomarkers able to monitor or predict a protective humoral immune response to the vaccine. In this review, we summarize these data, with emphasis on the effects of aging on influenza vaccine-specific B cell responses in healthy individuals and patients with Type-2 Diabetes, HIV and cardiovascular diseases.
Introduction
Vaccination has proven to be a very effective tool for preventing infectious disease [1]*. In general, vaccines elicit an immune response and consequently immunological memory that mediates protection from the infection. Influenza is associated with morbidity and mortality in very young and very old individuals as well as in individuals with immunodeficiency diseases, cardiovascular diseases, cerebrovascular disease, diabetes, chronic respiratory conditions and pregnancy [2]. The complications of influenza may include primary influenza pneumonia or secondary bacterial infections and exacerbations of pre-existing medical conditions. Hospitalization is the major contributor to the development of disability in elderly individuals [3,4]. The associated decline in physical activities and consequent disability represent a significant economic burden due to both direct (medical) and indirect costs (inability to work, reduction in productivity) [5]. Seasonal influenza epidemics are responsible for almost 200,000 estimated hospitalizations and 35,000 deaths each year in the United States and the elderly account for 90% of these 35,000 [6].
Vaccines against influenza require annual reformulation due to continuous viral evolution (antigenic drift and shift) which allows not only new human but also non-human influenza viruses to infect human beings. Annual influenza vaccinations help individuals to make protective antibodies specific for the currently circulating strains [7,8]. The influenza vaccine induces an antiviral response in B and T cells, resulting in humoral and cellular immunity, respectively [9]. The antibody response to the vaccine is the first line of protection from subsequent infection. An essential step in the generation of vaccine-induced antibody-secreting cells is the interaction of vaccine-specific B cells and T follicular helper cells (Tfh), to generate B cell proliferation, class switch recombination (CSR) and somatic hypermutation (SHM) [10].
It has been shown that some elderly individuals can still be infected with influenza even if they routinely receive the vaccine. This often leads to secondary complications, hospitalization, physical debilitation and ultimately death [11,12,13], likely due to a compromized immune system in these individuals. The fact that influenza vaccines also prevent complications from influenza (e.g. pneumonia) in most elderly strongly supports vaccination campaigns targeted to improve immune functions in these vulnerable individuals as will also be supported herein. Current influenza vaccination campaigns are able to reduce hospitalization to some extent [14], but rates of hospitalizations due to influenza-related disease are still very high [15].
The effects of influenza vaccination are different in individuals of different ages [16,17,18,19,20] and this depends on age-related differences in the innate and adaptive immune systems. These differences include a decrease in natural killer cell cytotoxicity on a per cell basis [21], a decrease in both numbers and function of dendritic cells in blood [22,23], a decrease in T cell function [24,25,26] and expression of CD28 [27], an increase in cytomegalovirus (CMV) seropositivity [28,29,30,31], and a decrease in B cell numbers and function [9,28,32,33,34,35], such as reduced CSR and SHM, leading to reduced generation of protective antibodies [35,36,37,38].
In this review we will summarize results on the effects of aging on influenza vaccine-specific B cell responses in healthy individuals as well as in individuals with Type-2 Diabetes (T2D), HIV and cardiovascular diseases (CVD).
Influenza vaccine-specific antibody responses in individuals of different age
Healthy individuals
Aging significantly decreases the influenza vaccine-specific antibody response in healthy individuals as we [36,37,38] and others have shown [9,17,39,40]. Most of the studies conducted so far have shown that this correlates with the well characterized age-dependent decrease in T cell [26,41,42] and dendritic cell [23] function. For T cells in particular, a shift with aging toward an anti-inflammatory response characterized by IL-10 production and decreased IFN-γ:IL-10 ratio in influenza-stimulated lymphocytes has been shown to be associated with reduced cytolytic capacity of CD8+ T cells which clear influenza virus from infected lungs [43]. However, we have shown that age-related intrinsic B cell defects also occur in blood and these contribute to decreased vaccine response. These include decreases in class switch recombination (CSR), the process that generates protective antibodies and memory B cells; decreases in the expression of the enzyme, activation-induced cytidine deaminase (AID), the transcription factor E47, which contributes to AID regulation; and decreased percentages of switched memory B cells (CD19+CD27+IgD-) before and after vaccination as compared with younger individuals.
We have measured the antibody response to the influenza vaccine in sera (in vivo response) and have associated this with the B cell response after vaccination to the vaccine in vitro. In vivo and in vitro B cell responses have been measured respectively by hemagglutination inhibition assay (HAI) and by AID mRNA expression by qPCR after B cell restimulation. AID is a measure of CSR and of B cell function which we have previously established to reflect the generation of specific IgG and associate with other mechanistic markers such as E47 [44]. Our results have shown that the specific response of B cells to vaccination in vivo and in vitro are both decreased with age and are significantly correlated [36,37,38]. These results support our hypothesis that the in vitro AID response recapitulates what has occurred in vivo in the germinal center in the generation of memory B cells. We have also shown that switched memory B cell percentages are good B cell biomarkers that are reduced in the blood of elderly individuals and are significantly correlated with the in vivo antibody response to the vaccine [36,37].
We have also demonstrated that CpG-induced AID at t0 and the percentage/number of switched memory B cells at t0 predict the ability to respond well to the influenza vaccine. Therefore, we suggest that AID in stimulated B cells and switched memory B cells at t0 are good predictive biomarkers for response to vaccines and infectious agents in humans [36,37]. More recently, we have demonstrated that unstimulated B cells make intracellular TNF-α and the levels of this pro-inflammatory cytokine are negatively correlated with the antibody response to the vaccine [45]. These 3 predictive biomarkers we have identified correlate with optimal antibody responses to the influenza vaccine in the majority of individuals (Figure 1). We have confirmed and extended these results showing for the first time a negative association between CMV-seropositivity and these B cell predictive biomarkers of optimal vaccine responses (Frasca et al., submitted). CMV seropositivity had also been shown to have a negative effect on influenza vaccine-specific antibody responses [39], due to accumulation of late-differentiated T cells (TEMRA).
Figure 1. B cell predictive biomarkers of in vivo influenza vaccine responses.
Left. AID mRNA expression (measured by qPCR) in CpG-stimulated B cells at t0 (before vaccination) predicts the in vivo response (as measured by fold-increase after vaccination) in 92% young (<60) and 74% elderly (≥60) individuals. Results are from 62 young and 39 elderly recruited during the 2011–2012 influenza vaccine season. Center. The percentages of switched memory (SM) B cells (CD27+IgD-) at t0 predict the in vivo response in 87% young and 79% elderly individuals (same in A). Right. Intracellular TNF-α (measured by flow cytometry) in unstimulated B cells at t0 predict a low in vivo response in 85% young and 73% elderly individuals. Results are from 33 young and 30 elderly recruited during the 2011–2012 influenza vaccine season.
The role of AID in CSR was well characterized previously [46,47] but its role in SHM and polyclonal antibody affinity maturation was only associated in those with genetic deficiency in mice [46] and humans [48] or weakly associated [49,50]. More recently, we have shown that AID correlates with high affinity antibodies specific for the pandemic (p)H1N1 influenza vaccine [38]. Our results showed that AID mRNA expression before vaccination in stimulated B cells as well as the fold-increase of AID mRNA expression after vaccination directly correlated with the increase in polyclonal antibody affinity to the HA1 globular domain of pH1N1 which is the domain most associated with protection to infection. In young individuals, significant affinity maturation to the HA1 globular domain was observed, which associated with initial levels of AID and fold-increase in AID after vaccination. In most of the elderly individuals, high affinity to the HA1 domain was observed before vaccination which resulted in minimal change in antibody affinity and correlated with low AID induction in this age group. In the elderly this likely reflects previous vaccination or infection with highly conserved pH1N1 determinants which generated an optimal memory response. These findings have demonstrated for the first time a strong correlation between AID induction and in vivo antibody affinity maturation in humans.
Others have measured the quality of antibodies secreted from single plasmablasts in young and elderly individuals 7 days after vaccination [40]. Results showed that not only the number of vaccine-specific plasmablasts decreased in elderly individuals, but also the amount of antibodies made by these cells is decreased. Conversely, the avidity of these vaccine-specific antibodies and the affinity of recombinant monoclonal antibodies obtained from single cell plasmablasts were similar in the two age groups. The authors reported that results support the conclusion that the lower efficacy of the influenza vaccine in the elderly is primarily due to a quantitative difference in the number of antibody-secreting plasmablasts which would be consistent with our data above showing optimal affinity towards cross-reactive or previously encountered antigens in the elderly already at t0 as well as after vaccination [38].
Also consistent with the above results, analyses of the clonal structure and mutation distribution of B cell repertoires in individuals of different ages vaccinated with the seasonal influenza vaccine have shown that elderly individuals had increased mutations before vaccination in their repertoires, suggesting that a priming by previous infections or vaccinations may have occurred [51]. Moreover, most of these elderly individuals show a reduced B cell clonal diversity as compared to young individuals. Their overall conclusion that the “higher numbers of somatic mutations observed earlier in elderly individuals arise from clonal expansions that draw upon a pool of B cells having more somatic mutations to begin with” agrees with the other results above [38,40].
Because the 3 antigens in the influenza vaccine were repeated in the 2010/11, 2011/12 seasons, possible defects in the antibody response in the elderly due to the deficits we have seen in AID would have been underestimated. We are currently measuring HAI responses and avidity to the new antigens present in the vaccine in the last 2 seasons.
Patients with T2D
T2D patients are at elevated risk for infections due to influenza or for complications related to it [52] and therefore annual vaccination is highly recommended in these patients. Viral and bacterial infections and consequent diseases in T2D patients have been shown to cause loss of metabolic control and consequent increase of glycosylated serum proteins, ketoacidosis which may result in an increased hospitalization, increased mortality rates and prolonged complications [53]. Not much is known about the effects of influenza vaccination on lymphocyte populations of T2D patients. A few studies have measured T cell function in vaccinated young [54] and elderly [55] T2D patients as compared to healthy controls and have shown reduced [54] or similar [55] responses in patients versus controls.
We have evaluated in vivo and in vitro B cell responses to the seasonal influenza vaccine in normal healthy controls and in T2D patients [56]. Briefly, we demonstrated that both in vivo and in vitro responses decrease by age in healthy individuals but not in T2D patients, despite high levels of serum and B cell-intrinsic inflammation (TNF-α) in T2D patients. This was surprising as we had previously demonstrated these negatively impact B cell function in mice [57] and humans [45]. We hypothesized that this may be due to the fact that the innate immune system of T2D patients is beneficially hyperactivated, as we found elevated serum levels of bacterial lipopolysaccharide (LPS) and soluble (s)CD14, and we suggest that these may not only counteract the negative effects of inflammation from increased TNF-α, IL-6 and CRP, but also to induce a direct stimulation of B cells. Another explanation for these data is that the T2D patients in our study were taking anti-inflammatory agents, such as Metformin, which is known to block TNF-α signaling in all cells, including B cells.
Patients with HIV
HIV viral infection has been shown to be associated with an aberrant activation of cells of the immune system, including B cells, which show phenotypic and functional alterations. B cells from HIV-infected patients show impaired reactivity to in vivo and in vitro activation [58,59]. Seasonal influenza vaccination is recommended for HIV-infected individuals to reduce influenza-related morbidity and mortality. HIV-infected people are at a significantly higher risk than the general population at all ages for influenza infection, despite vaccination and virologic control with combined antiretroviral therapy (cART) [60,61]. However, the response to influenza vaccination is frequently impaired in these individuals [62,63]. Intrinsic defects in B cells have been described in chronically HIV-positive individuals [64]. In addition, HIV infection has been shown to have a crucial role in the cellular senescence and immunodeficiency of both T and B cells [65,66].
In order to investigate the cumulative effects of HIV infection and aging on the response to the seasonal influenza vaccine, we recently performed a study in which we enrolled HIV-infected post-menopausal women undergoing cART and healthy controls [59]. We found that the antibody responses to the influenza vaccine were lower in HIV-infected as compared to uninfected controls. Moreover, HIV-infected women had higher frequencies of activated, proliferating CD4 and Tfh cells (CD38+HLADR-Ki67+) and of activated, senescent CD8 T cells (CD8+CD28−). These were negatively correlated with antibody titers after vaccination (also in the HIV-uninfected group). High pre-vaccination levels of TNF-α in plasma were found to be associated with Tfh activation and low antibody responses to the influenza vaccine.
Patients with CVD
Immunization against seasonal influenza has a critical role in preventing morbidity and mortality in patients with CVD. The American Heart Association and the American College of Cardiology recommend influenza vaccination as part of comprehensive secondary prevention in children, adults and elderly individuals with coronary and other atherosclerotic vascular diseases [67]. Nevertheless, influenza vaccination coverage levels among persons with CVD remain below national goals and influenza-related death is more common among individuals with CVD than among patients with any other chronic condition [68,69]. This situation is exacerbated in the elderly in which the dysregulation of the inflammatory network due to various age-associated chronic stresses also occur, increasing the susceptibility of these individuals to develop CVD [70].
There are limited references on the effects of influenza vaccination on lymphocyte subpopulations in CVD patients and the few studies performed have evaluated in vivo antibody production to the vaccine and associated this with T cell function. In one of these studies, the relationship among the development of influenza, serum antibody titers and ex vivo cellular immune responses to influenza vaccination in community dwelling elderly (60–90 years of age) including those with congestive heart failure (CHF) has been evaluated [71]. Results indicate that both healthy and CHF elderly showed seroconversion after vaccination, although the CHF patients had a lower titer as compared to healthy young adults. In addition, in healthy but not in CHF elderly individuals, T cell cellular responses (granzyme B levels) increased after in vitro stimulation of PBMC with the influenza virus. Another study also showed that patients with CHF have decreased T cell responses to the influenza virus, as compared to healthy controls, despite similar rates of seroprotection and seroconversion [72]. These apparently discrepant results may be due to different individuals and/or different degrees of pre-immunization.
Conclusions
Vaccination, and in particular the influenza vaccine, protects the majority from severe health complications associated with infection. Those populations especially susceptible include the elderly and those with co-morbidities such as T2D, HIV, CVD, as well as cancer and autoimmunity. Autonomous B lymphocyte deficiencies have been found which contribute to lower vaccine response in the elderly as well as in some young individuals and these should also be predictive in other risk groups. These include lower switched memory B cells, higer intracellular TNF-α in unstimulated B cells, and lower AID in stimulated B cells before vaccination. The basic science and clinical communities are increasingly aware and have contributed important new information to identify deficits in particular lymphocytic (T, B, NK) and myelocytic (monocytes, dendritic cells) subpopulations. Improvement in future vaccines will depend upon identifying and correcting deficits in multiple cell types to achieve optimal protective immune responses.
Highlights.
B cell defects increase with age and contribute to lower influenza vaccine response
Defects include lower Ig class switch recombination (CSR), lower activation-induced cytidine deaminase (AID), and higher serum/B cell TNF-α
T2D patients have high inflammation but do not have decreased vaccine response
HIV patients have high serum TNF-α, activated Tfh and lower vaccine response
Elderly CVD patients have decreased vaccine response
Acknowledgements
Supported by NIH grants AG032576, AG023717, AI096446, AG042826.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
The authors regret that they could not include more of the relevant references to this field. We have tried to reference articles we thought were historically significant and/or recently published.
References and recommended reading
- 1. Finco O, Rappuoli R. Designing Vaccines for the Twenty-First Century Society. Front Immunol. 2014;5:12. doi: 10.3389/fimmu.2014.00012. *This is a very comprehensive, interesting, and up-to-date review of the history, challeges and successes of vaccine development and suggestions for the future.
- 2.Yung CF, Andrews N, Hoschler K, Miller E. Comparing the immunogenicity of AS03-adjuvanted 2009 pandemic H1N1 vaccine with clinical protection in priority risk groups in England. PLoS One. 2013;8:e56844. doi: 10.1371/journal.pone.0056844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ferrucci L, Guralnik JM, Pahor M, Corti MC, Havlik RJ. Hospital diagnoses, Medicare charges, and nursing home admissions in the year when older persons become severely disabled. JAMA. 1997;277:728–734. [PubMed] [Google Scholar]
- 4.Pilotto A, Panza F, Sancarlo D, Paroni G, Maggi S, Ferrucci L. Usefulness of the multidimensional prognostic index (MPI) in the management of older patients with chronic kidney disease. J Nephrol. 2012;25(Suppl 19):S79–S84. doi: 10.5301/jn.5000162. [DOI] [PubMed] [Google Scholar]
- 5.Monto AS, Ansaldi F, Aspinall R, McElhaney JE, Montano LF, Nichol KL, Puig-Barbera J, Schmitt J, Stephenson I. Influenza control in the 21st century: Optimizing protection of older adults. Vaccine. 2009;27:5043–5053. doi: 10.1016/j.vaccine.2009.06.032. [DOI] [PubMed] [Google Scholar]
- 6.Thompson WW, Shay DK, Weintraub E, Brammer L, Cox N, Anderson LJ, Fukuda K. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA. 2003;289:179–186. doi: 10.1001/jama.289.2.179. [DOI] [PubMed] [Google Scholar]
- 7.McMurry JA, Johansson BE, De Groot AS. A call to cellular & humoral arms: enlisting cognate T cell help to develop broad-spectrum vaccines against influenza A. Hum Vaccin. 2008;4:148–157. doi: 10.4161/hv.4.2.5169. [DOI] [PubMed] [Google Scholar]
- 8.Wrammert J, Smith K, Miller J, Langley WA, Kokko K, Larsen C, Zheng NY, Mays I, Garman L, Helms C, et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature. 2008;453:667–671. doi: 10.1038/nature06890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Shi Y, Yamazaki T, Okubo Y, Uehara Y, Sugane K, Agematsu K. Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell. J Immunol. 2005;175:3262–3267. doi: 10.4049/jimmunol.175.5.3262. [DOI] [PubMed] [Google Scholar]
- 10.Crotty S. Follicular helper CD4 T cells (TFH) Annu Rev Immunol. 2011;29:621–663. doi: 10.1146/annurev-immunol-031210-101400. [DOI] [PubMed] [Google Scholar]
- 11.Gross PA, Hermogenes AW, Sacks HS, Lau J, Levandowski RA. The efficacy of influenza vaccine in elderly persons. A meta-analysis and review of the literature. Ann Intern Med. 1995;123:518–527. doi: 10.7326/0003-4819-123-7-199510010-00008. [DOI] [PubMed] [Google Scholar]
- 12.Simonsen L, Clarke MJ, Schonberger LB, Arden NH, Cox NJ, Fukuda K. Pandemic versus epidemic influenza mortality: a pattern of changing age distribution. J Infect Dis. 1998;178:53–60. doi: 10.1086/515616. [DOI] [PubMed] [Google Scholar]
- 13.Vu T, Farish S, Jenkins M, Kelly H. A meta-analysis of effectiveness of influenza vaccine in persons aged 65 years and over living in the community. Vaccine. 2002;20:1831–1836. doi: 10.1016/s0264-410x(02)00041-5. [DOI] [PubMed] [Google Scholar]
- 14.Nichol KL, Margolis KL, Wuorenma J, Von Sternberg T. The efficacy and cost effectiveness of vaccination against influenza among elderly persons living in the community. N Engl J Med. 1994;331:778–784. doi: 10.1056/NEJM199409223311206. [DOI] [PubMed] [Google Scholar]
- 15.Thompson WW, Shay DK, Weintraub E, Brammer L, Bridges CB, Cox NJ, Fukuda K. Influenza-associated hospitalizations in the United States. JAMA. 2004;292:1333–1340. doi: 10.1001/jama.292.11.1333. [DOI] [PubMed] [Google Scholar]
- 16.Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine. 2006;24:1159–1169. doi: 10.1016/j.vaccine.2005.08.105. [DOI] [PubMed] [Google Scholar]
- 17.McElhaney JE. Influenza vaccine responses in older adults. Ageing Res Rev. 2011;10:379–388. doi: 10.1016/j.arr.2010.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Colonna-Romano G, Bulati M, Aquino A, Scialabba G, Candore G, Lio D, Motta M, Malaguarnera M, Caruso C. B cells in the aged: CD27, CD5, and CD40 expression. Mech Ageing Dev. 2003;124:389–393. doi: 10.1016/s0047-6374(03)00013-7. [DOI] [PubMed] [Google Scholar]
- 19.Sambhara S, McElhaney JE. Immunosenescence and influenza vaccine efficacy. Curr Top Microbiol Immunol. 2009;333:413–429. doi: 10.1007/978-3-540-92165-3_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Simonsen L, Taylor RJ, Viboud C, Miller MA, Jackson LA. Mortality benefits of influenza vaccination in elderly people: an ongoing controversy. Lancet Infect Dis. 2007;7:658–666. doi: 10.1016/S1473-3099(07)70236-0. [DOI] [PubMed] [Google Scholar]
- 21.Solana R, Pawelec G, Tarazona R. Aging and innate immunity. Immunity. 2006;24:491–494. doi: 10.1016/j.immuni.2006.05.003. [DOI] [PubMed] [Google Scholar]
- 22.Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum Immunol. 2009;70:777–784. doi: 10.1016/j.humimm.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184:2518–2527. doi: 10.4049/jimmunol.0901022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Goronzy JJ, Lee WW, Weyand CM. Aging and T-cell diversity. Exp Gerontol. 2007;42:400–406. doi: 10.1016/j.exger.2006.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gupta S, Bi R, Su K, Yel L, Chiplunkar S, Gollapudi S. Characterization of naive, memory and effector CD8+ T cells: effect of age. Exp Gerontol. 2004;39:545–550. doi: 10.1016/j.exger.2003.08.013. [DOI] [PubMed] [Google Scholar]
- 26.Pawelec G, Barnett Y, Forsey R, Frasca D, Globerson A, McLeod J, Caruso C, Franceschi C, Fulop T, Gupta S, et al. T cells and aging, January 2002 update. Front Biosci. 2002;7:d1056–d1183. doi: 10.2741/a831. [DOI] [PubMed] [Google Scholar]
- 27.Vallejo AN. CD28 extinction in human T cells: altered functions and the program of T-cell senescence. Immunol Rev. 2005;205:158–169. doi: 10.1111/j.0105-2896.2005.00256.x. [DOI] [PubMed] [Google Scholar]
- 28.Gibson KL, Wu YC, Barnett Y, Duggan O, Vaughan R, Kondeatis E, Nilsson BO, Wikby A, Kipling D, Dunn-Walters DK. B-cell diversity decreases in old age and is correlated with poor health status. Aging Cell. 2009;8:18–25. doi: 10.1111/j.1474-9726.2008.00443.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Olsson J, Wikby A, Johansson B, Lofgren S, Nilsson BO, Ferguson FG. Age-related change in peripheral blood T-lymphocyte subpopulations and cytomegalovirus infection in the very old: the Swedish longitudinal OCTO immune study. Mech Ageing Dev. 2000;121:187–201. doi: 10.1016/s0047-6374(00)00210-4. [DOI] [PubMed] [Google Scholar]
- 30.Pawelec G, Akbar A, Caruso C, Solana R, Grubeck-Loebenstein B, Wikby A. Human immunosenescence: is it infectious? Immunol Rev. 2005;205:257–268. doi: 10.1111/j.0105-2896.2005.00271.x. [DOI] [PubMed] [Google Scholar]
- 31.Pawelec G, Derhovanessian E, Larbi A, Strindhall J, Wikby A. Cytomegalovirus and human immunosenescence. Rev Med Virol. 2009;19:47–56. doi: 10.1002/rmv.598. [DOI] [PubMed] [Google Scholar]
- 32.Ademokun A, Wu YC, Dunn-Walters D. The ageing B cell population: composition and function. Biogerontology. 2010;11:125–137. doi: 10.1007/s10522-009-9256-9. [DOI] [PubMed] [Google Scholar]
- 33.Buffa S, Pellicano M, Bulati M, Martorana A, Goldeck D, Caruso C, Pawelec G, Colonna-Romano G. A novel B cell population revealed by a CD38/CD24 gating strategy: CD38(-)CD24 (-) B cells in centenarian offspring and elderly people. Age (Dordr) 2013;35:2009–2024. doi: 10.1007/s11357-012-9488-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chong Y, Ikematsu H, Yamaji K, Nishimura M, Nabeshima S, Kashiwagi S, Hayashi J. CD27(+) (memory) B cell decrease and apoptosis-resistant CD27(-) (naive) B cell increase in aged humans: implications for age-related peripheral B cell developmental disturbances. Int Immunol. 2005;17:383–390. doi: 10.1093/intimm/dxh218. [DOI] [PubMed] [Google Scholar]
- 35.Frasca D, Landin AM, Lechner SC, Ryan JG, Schwartz R, Riley RL, Blomberg BB. Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and Ig class switch in human B cells. J Immunol. 2008;180:5283–5290. doi: 10.4049/jimmunol.180.8.5283. [DOI] [PubMed] [Google Scholar]
- 36. Frasca D, Diaz A, Romero M, Landin AM, Phillips M, Lechner SC, Ryan JG, Blomberg BB. Intrinsic defects in B cell response to seasonal influenza vaccination in elderly humans. Vaccine. 2010;28:8077–8084. doi: 10.1016/j.vaccine.2010.10.023. ** This paper was the first to show that intrinsic B cell defects contribute to a reduced antibody vaccine response in elderly humans.
- 37.Frasca D, Diaz A, Romero M, Phillips M, Mendez NV, Landin AM, Blomberg BB. Unique biomarkers for B-cell function predict the serum response to pandemic H1N1 influenza vaccine. Int Immunol. 2012;24:175–182. doi: 10.1093/intimm/dxr123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Khurana S, Frasca D, Blomberg B, Golding H. AID activity in B cells strongly correlates with polyclonal antibody affinity maturation in-vivo following pandemic 2009-H1N1 vaccination in humans. PLoS Pathog. 2012;8:e1002920. doi: 10.1371/journal.ppat.1002920. **AID was shown for the first time to be correlated with antibody affinity to the influenza vaccine in humans.
- 39. Derhovanessian E, Theeten H, Hahnel K, Van Damme P, Cools N, Pawelec G. Cytomegalovirus-associated accumulation of late-differentiated CD4 T-cells correlates with poor humoral response to influenza vaccination. Vaccine. 2013;31:685–690. doi: 10.1016/j.vaccine.2012.11.041. * Latent CMV infection associated with lower response to the intradermal vaccine, Intanza, in the elderly.
- 40.Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, Nishtala M, Wrammert J, Smith K, James JA, et al. Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with decreased production of vaccine-specific antibodies. J Clin Invest. 2011;121:3109–3119. doi: 10.1172/JCI57834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Goronzy JJ, Fulbright JW, Crowson CS, Poland GA, O'Fallon WM, Weyand CM. Value of immunological markers in predicting responsiveness to influenza vaccination in elderly individuals. J Virol. 2001;75:12182–12187. doi: 10.1128/JVI.75.24.12182-12187.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Saurwein-Teissl M, Lung TL, Marx F, Gschosser C, Asch E, Blasko I, Parson W, Bock G, Schonitzer D, Trannoy E, et al. Lack of antibody production following immunization in old age: association with CD8(+)CD28(−) T cell clonal expansions and an imbalance in the production of Th1 and Th2 cytokines. J Immunol. 2002;168:5893–5899. doi: 10.4049/jimmunol.168.11.5893. [DOI] [PubMed] [Google Scholar]
- 43. McElhaney JE, Zhou X, Talbot HK, Soethout E, Bleackley RC, Granville DJ, Pawelec G. The unmet need in the elderly: how immunosenescence, CMV infection, co-morbidities and frailty are a challenge for the development of more effective influenza vaccines. Vaccine. 2012;30:2060–2067. doi: 10.1016/j.vaccine.2012.01.015. **CMV-infected, late stage CD8 T cells at the infection site are not cytotoxic and also suppressive via granzyme B.
- 44.Frasca D, Landin AM, Alvarez JP, Blackshear PJ, Riley RL, Blomberg BB. Tristetraprolin, a negative regulator of mRNA stability, is increased in old B cells and is involved in the degradation of E47 mRNA. J Immunol. 2007;179:918–927. doi: 10.4049/jimmunol.179.2.918. [DOI] [PubMed] [Google Scholar]
- 45.Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. High TNF-alpha levels in resting B cells negatively correlate with their response. Exp Gerontol. 2014;54:116–122. doi: 10.1016/j.exger.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, Honjo T. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell. 2000;102:553–563. doi: 10.1016/s0092-8674(00)00078-7. [DOI] [PubMed] [Google Scholar]
- 47.Muramatsu M, Nagaoka H, Shinkura R, Begum NA, Honjo T. Discovery of activation-induced cytidine deaminase, the engraver of antibody memory. Adv Immunol. 2007;94:1–36. doi: 10.1016/S0065-2776(06)94001-2. [DOI] [PubMed] [Google Scholar]
- 48.Durandy A. Hyper-IgM syndromes: a model for studying the regulation of class switch recombination and somatic hypermutation generation. Biochem Soc Trans. 2002;30:815–818. doi: 10.1042/bst0300815. [DOI] [PubMed] [Google Scholar]
- 49.Dorsett Y, McBride KM, Jankovic M, Gazumyan A, Thai TH, Robbiani DF, Di Virgilio M, Reina San-Martin B, Heidkamp G, Schwickert TA, et al. MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity. 2008;28:630–638. doi: 10.1016/j.immuni.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Teng G, Hakimpour P, Landgraf P, Rice A, Tuschl T, Casellas R, Papavasiliou FN. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity. 2008;28:621–629. doi: 10.1016/j.immuni.2008.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jiang N, He J, Weinstein JA, Penland L, Sasaki S, He XS, Dekker CL, Zheng NY, Huang M, Sullivan M, et al. Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci Transl Med. 2013;5:171ra119. doi: 10.1126/scitranslmed.3004794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Muller LM, Gorter KJ, Hak E, Goudzwaard WL, Schellevis FG, Hoepelman AI, Rutten GE. Increased risk of common infections in patients with type 1 and type 2 diabetes mellitus. Clin Infect Dis. 2005;41:281–288. doi: 10.1086/431587. [DOI] [PubMed] [Google Scholar]
- 53.Eickhoff TC, Sherman IL, Serfling RE. Observations on excess mortality associated with epidemic influenza. JAMA. 1961;176:776–782. doi: 10.1001/jama.1961.03040220024005. [DOI] [PubMed] [Google Scholar]
- 54.Pozzilli P, Gale EA, Visalli N, Baroni M, Crovari P, Frighi V, Cavallo MG, Andreani D. The immune response to influenza vaccination in diabetic patients. Diabetologia. 1986;29:850–854. doi: 10.1007/BF00870139. [DOI] [PubMed] [Google Scholar]
- 55.McElhaney JE, Pinkoski MJ, Au D, Lechelt KE, Bleackley RC, Meneilly GS. Helper and cytotoxic T lymphocyte responses to influenza vaccination in healthy compared to diabetic elderly. Vaccine. 1996;14:539–544. doi: 10.1016/0264-410x(95)00219-q. [DOI] [PubMed] [Google Scholar]
- 56. Frasca D, Diaz A, Romero M, Mendez NV, Landin AM, Ryan JG, Blomberg BB. Young and elderly patients with type 2 diabetes have optimal B cell responses to the seasonal influenza vaccine. Vaccine. 2013;31:3603–3610. doi: 10.1016/j.vaccine.2013.05.003. *Unexpectedly,T2D patients with high levels of B-cell instrinsic TNF-α, responded well to the vaccine, alternative reasons are propsed.
- 57.Frasca D, Romero M, Diaz A, Alter-Wolf S, Ratliff M, Landin AM, Riley RL, Blomberg BB. A molecular mechanism for TNF-alpha-mediated downregulation of B cell responses. J Immunol. 2012;188:279–286. doi: 10.4049/jimmunol.1003964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Moir S, Fauci AS. Pathogenic mechanisms of B-lymphocyte dysfunction in HIV disease. J Allergy Clin Immunol. 2008;122:12–19. doi: 10.1016/j.jaci.2008.04.034. quiz 20-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Parmigiani A, Alcaide ML, Freguja R, Pallikkuth S, Frasca D, Fischl MA, Pahwa S. Impaired antibody response to influenza vaccine in HIV-infected and uninfected aging women is associated with immune activation and inflammation. PLoS One. 2013;8:e79816. doi: 10.1371/journal.pone.0079816. *Plasma IL-21 and TNF-α were positively and negatively respectively correlated with seroconversion to the vaccine. TNF-α correlated with activated pTfh cells.
- 60.Cohen C, Simonsen L, Sample J, Kang JW, Miller M, Madhi SA, Campsmith M, Viboud C. Influenza-related mortality among adults aged 25–54 years with AIDS in South Africa and the United States of America. Clin Infect Dis. 2012;55:996–1003. doi: 10.1093/cid/cis549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sheth AN, Patel P, Peters PJ. Influenza and HIV: lessons from the 2009 H1N1 influenza pandemic. Curr HIV/AIDS Rep. 2011;8:181–191. doi: 10.1007/s11904-011-0086-4. [DOI] [PubMed] [Google Scholar]
- 62.Malaspina A, Moir S, Orsega SM, Vasquez J, Miller NJ, Donoghue ET, Kottilil S, Gezmu M, Follmann D, Vodeiko GM, et al. Compromised B cell responses to influenza vaccination in HIV-infected individuals. J Infect Dis. 2005;191:1442–1450. doi: 10.1086/429298. [DOI] [PubMed] [Google Scholar]
- 63.Tebas P, Frank I, Lewis M, Quinn J, Zifchak L, Thomas A, Kenney T, Kappes R, Wagner W, Maffei K, et al. Poor immunogenicity of the H1N1 2009 vaccine in well controlled HIV-infected individuals. AIDS. 2010;24:2187–2192. doi: 10.1097/QAD.0b013e32833c6d5c. [DOI] [PubMed] [Google Scholar]
- 64. Cagigi A, Pensieroso S, Ruffin N, Sammicheli S, Thorstensson R, Pan-Hammarstrom Q, Hejdeman B, Nilsson A, Chiodi F. Relation of activation-induced deaminase (AID) expression with antibody response to A(H1N1)pdm09 vaccination in HIV-1 infected patients. Vaccine. 2013;31:2231–2237. doi: 10.1016/j.vaccine.2013.03.002. *AID expression in blood B-cells predicted the antibody response to influenza vaccine in HIV-1 infected patients with similar CD4+ T cell counts and age.
- 65.Chou JP, Ramirez CM, Wu JE, Effros RB. Accelerated aging in HIV/AIDS: novel biomarkers of senescent human CD8+ T cells. PLoS One. 2013;8:e64702. doi: 10.1371/journal.pone.0064702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pommier JP, Gauthier L, Livartowski J, Galanaud P, Boue F, Dulioust A, Marce D, Ducray C, Sabatier L, Lebeau J, et al. Immunosenescence in HIV pathogenesis. Virology. 1997;231:148–154. doi: 10.1006/viro.1997.8512. [DOI] [PubMed] [Google Scholar]
- 67.Smith SC, Jr, Allen J, Blair SN, Bonow RO, Brass LM, Fonarow GC, Grundy SM, Hiratzka L, Jones D, Krumholz HM, et al. AHA/ACC guidelines for secondary prevention for patients with coronary and other atherosclerotic vascular disease: 2006 update: endorsed by the National Heart, Lung, and Blood Institute. Circulation. 2006;113:2363–2372. doi: 10.1161/CIRCULATIONAHA.106.174516. [DOI] [PubMed] [Google Scholar]
- 68.Douglas RG, Jr, Betts RF, Simons RL, Hogan PW, Roth FK. Evaluation of a topical interferon inducer in experimental influenza infection in volunteers. Antimicrob Agents Chemother. 1975;8:684–687. doi: 10.1128/aac.8.6.684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Glezen WP, Decker M, Perrotta DM. Survey of underlying conditions of persons hospitalized with acute respiratory disease during influenza epidemics in Houston, 1978–1981. Am Rev Respir Dis. 1987;136:550–555. doi: 10.1164/ajrccm/136.3.550. [DOI] [PubMed] [Google Scholar]
- 70.De Martinis M, Franceschi C, Monti D, Ginaldi L. Inflamm-ageing and lifelong antigenic load as major determinants of ageing rate and longevity. FEBS Lett. 2005;579:2035–2039. doi: 10.1016/j.febslet.2005.02.055. [DOI] [PubMed] [Google Scholar]
- 71.McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, Ewen C, Kane KP, Bleackley RC. T cell responses are better correlates of vaccine protection in the elderly. J Immunol. 2006;176:6333–6339. doi: 10.4049/jimmunol.176.10.6333. [DOI] [PubMed] [Google Scholar]
- 72.Vardeny O, Moran JJ, Sweitzer NK, Johnson MR, Hayney MS. Decreased T-cell responses to influenza vaccination in patients with heart failure. Pharmacotherapy. 2010;30:10–16. doi: 10.1592/phco.30.1.10. [DOI] [PMC free article] [PubMed] [Google Scholar]