Abstract
The immune system undergoes a complex and continuous remodeling with aging. Immunosenescence results into both quantitative and qualitative changes of specific cellular subpopulations that have major impact on allorecognition and alloresponse, and consequently on graft rejection and tolerance. Here, we are going to review the immunological changes associated with the aging process relevant for transplantation. Interventions to selectively target changes associated with the senescence process seem promising therapeutic strategies to improve transplantation outcome.
Keywords: aging, elderly, immunosenescence, transplantation
INTRODUCTION
The number of older people is increasing dramatically. In the world, by year 2050 the number of older people will surpass the number of younger (<35 years of age) people for the first time [1]. The exponential increasing proportion of elderly individuals in our society and the shortage of organs impose significant challenges to organ transplantation.
The elderly is the fastest growing segment of the population with end-stage organ disease [2]. In the United States, transplant recipients of any solid organ older than 65 years represented in 2012 9.3% of all recipients, while in 1988 they represented only 2.1%. Likewise, the percentage of solid organ donors older than 65 years increased more than 8 times in the same period, representing 7.1% in 2012 [2].
Immunosenescence is the process of progressive dysfunction of the immune system that increases the susceptibility of the elderly to infection, autoimmune disease, and cancer, contributing significantly to morbidity and mortality of the elderly [3–5].
Here, we are going to update the literature and discuss the complex remodeling of the immune system in the elderly by describing most significant changes of key players, and its impact on both rejection and tolerance.
CHANGES OF THE IMMUNE SYSTEM WITH AGING
The aging process causes important anatomical and functional changes in a number of systems that result in reduction of the functional reserve and inability to cope with stress. As the functional reserve decreases, organs are exposed to overload during stress conditions, resulting in a vicious cycle activating the immune system while contributing to tissue injury and reduction of function [6, 7].
The immune system undergoes a complex and continuous remodeling with aging. Immunosenescence changes result into both quantitative and qualitative modifications of specific cellular subpopulations rather than a global deterioration of the immune system, as previously thought [8, 9]. Even stem cells, despite their extensive proliferative and regenerative capacity, show signs of aging [10, 11].
The most striking alterations are found in phenotypes and functions of T-cell components and less frequently in components of the natural (innate) immune system. Consequently, chemotaxis, phagocytosis, natural cytotoxicity, and complement activity are relatively well preserved in elderly individuals [12]. Most alterations of B cells seem secondary to T-cell dysfunction [4].
The most relevant age-related immunological modifications are on T cells, since they play a pivotal role in tolerance and rejection [13]. Regarding the changes in the absolute number of lymphocytes with aging there is some controversy [14], whereas there is sufficient evidence for an alteration of specific cell subsets. Changes in T-cell subtypes include increased amounts of CD3+, CD4+, CD8+ T cells, decreased CD28+ T cells, and a shift from naïve to memory phenotypes, leading to a limited T-cell repertoire [3, 9, 12]. With aging, the diversity of TCR-b chains drops 1 000-fold in individuals older than 70 years [15].
It is hypothesized that aging is associated with chronic activation of the immune system [16]. The cytokine network undergoes profound and complex changes with aging [17]. It has been shown that there is a shift toward the production of pro-inflammatory cytokines (IL-6, IL-1β, TNF-α, and IFN-γ), and reduction of the expression of several adhesion molecules and chemokine receptor expression with increasing age [18]. In addition, aging of the endocrine and neurological systems are closely associated with aging of the immunological system and may contribute to chronic allograft deterioration [18, 19].
Aged Memory Cells
The number of memory cells increases with age, possibly reflecting an accumulation of antigenic experiences. In neonates, 99% of T-cells are naïve, while in the age group from 50 to 70 years only 35% are naive, and in centenarians about 20% [12, 20]. Alloreactive memory cells, generated either by previous sensitization to alloantigens or through heterologous immunity, are barriers to achieve graft tolerance [20]. Unlike naïve T-cells, memory cells can be fully activated in the absence of costimulation [21, 22], can recirculate in peripheral nonlymphoid tissues, and be able to initiate early responses directly in the graft. Memory cells response to antigens has greater magnitude and efficacy than response from naïve T cells [23]. It has been shown that a high number of memory cells are associated with increased incidence and severity of rejection [24].
With aging, the number of immature B cells decrease and there is a significant loss in diversity of the B-cell receptor [25, 26]. The numbers of memory B cells (CD27+) are increased with aging [27]. An increased proportion of mature B cells may also account for the increased resistance to tolerance induction in the elderly [28–31].
MHC expression in aged cells
MHC class II plays a key role in regulating and restricting the immune response [32–34]. Optimal levels of MHC expression are crucial for the proper functioning of cellular and humoral immune responses. MHC class I protein levels increase significantly with age on both peripheral blood and spleen (T cells and B cells) lymphocytes, whereas the percentage of MHC class II-expressing spleen lymphocytes markedly decreases [35–37]. This decrease was due to a decrease in the relative proportion of B cells compared to T cells in the spleen lymphocyte population of older animals [35]. The ease of B-cell tolerance induction decreases with aging in native, and lethally irradiated and thymectomized mice recipients of B cells from donors of different ages. This age resistance of the peripheral B-cell population to tolerance induction might be, at least in part, accounted for the increased incidence of auto-antibodies [38, 39].
Aged T-regulatory cells
Regulatory T cells (Tregs) play crucial roles in the induction and maintenance of allograft tolerance. There is growing evidence indicating that Tregs control the activation of primary and memory T-cell responses. The literature on changes of Treg with aging is limited and controversial [40]. There are studies showing both increase and decrease in the numbers of Tregs [41–45]. It has been shown in aged porcine thymi that there are fewer regulatory cells to inhibit alloreactive cells [42]. Regarding Treg function, most studies showed that aged Tregs are dysfunctional and unable to suppress aged T-effector cells. In a mouse model, one study indicated that the percentages, phenotypes, the size of TCR repertoire, and function of CD4+ CD25+ Treg cells were altered significantly with aging. It showed that CD4+ CD25+ Treg cells of aged mice displayed significantly lower inhibiting ability on alloantigen-induced delayed-type hypersensitivity reaction or on inflammatory cytokines (IL-2 and IFN-gamma) production, but not on cell proliferation of effector T cells [43]. In addition, there was a direct correlation between the expansion of Treg cells and the aged immune deficiency, and the authors speculated that depletion of these cells might be critical for restoring immune responses in aged animals [44]. In a clinical study, it was found that Tregs, accumulated as a result of aging, were capable of suppressing cytotoxic activity of CD8+ T and NK cells, and production of IL-2 [45].
It has been shown that Tregs in aged individuals may have a restricted TCR repertoire [46]. Studies in humans demonstrated that the suppressive activity of CD4+CD25+ Tregs declines with age [47]. Other study showed that thymocytes of aged mice have a significant increase in the percentage of CD4+ CD25+ cells than in young mice [48]. In this study, the expression of surface markers also changed with aging. CD4+ CD25+ cell expression of CD69, CD5, CD28, and FoxP3 was lower, whereas the expression of CTLA-4 and CD28 was higher. Invitro studies showed that these aged CD4+ CD25+ cells maintained their potential to suppress the proliferation of activated responder lymphocytes of young mice, but not the proliferation of responder T cells from aged animals, implying that the response dysfunction may lie in changes of the CD4+ CD25− effector T-cell population’s ability to proliferate or respond to Tregs [48, 49].
Dendritic cells and costimulatory pathways in the elderly
Dendritic cells (DCs) are the most effective antigen presenting cells to initiate the immune response and play an important role on both rejection and tolerance, depending on the graft microenvironment [50]. Costimulatory signals, originating exclusively from professional antigen presenting cells, set the stage for either rejection or tolerance. Through “positive” costimulatory molecules and “negative” T-cell costimulatory pathways, the function of the immune system can be activated or down-regulated, respectively. In the elderly, the costimulatory system is dysfunctional [51]. It has also been shown that aging DCs have impaired migration to draining lymph nodes [52]. Data on age-associated changes in the number and function of DCs are controversial and vary depending on the subset of DCs and in the compartment in which they are located [52]. DC function is increased in healthy aged humans and mice, and may enhance allorecognition to compensate for an impaired function of senescent T cells [53–55]. However, in frail elderly patient, DC function deteriorates, reducing antigen presentation, expression of costimulatory molecules, and IL-2 production [16].
Della Bella et al. observed that DCs from aged individual have a more mature phenotype with higher expression of costimulatory molecules CD86 and CD83 [56]. Varas et al. observed a decrease in the density of thymic stromal DC with aging. They found reduced expression of MHC-II, CD40, CD86, and CD54 in thymic DCs from aged mice along with decreased allostimulatory capacity [57]. These stromal thymic DCs play a critical role in selection of Tregs and might explain differences in the subset of Tregs and resistance to tolerance. In accordance to this hypothesis, another study showed that different expression of CD80, CD86, and CD40 on DCs of old mice may explain the elevation of the percentage of natural Tregs in the thymus with aging [58].
With aging, the CTLA4 molecule is increased [59], whereas CD28, CD40-CD40L, MHC class II expression are reduced [35, 60–62]. At birth, CD28 costimulatory molecule is expressed on more than 99% of T cells, and at ages 70 to 90 it is reduced to 71% [60].
Innate immune system in the elderly
There is increasing data on the importance of the innate immune response in both rejection and tolerance [63–69]. All of components of the innate immune system are also affected by advanced age [70–73]. There is evidence that aging is associated with a hyperinflammatory state that may exacerbate chronic conditions such as atherosclerosis. [74]. There is an increase in pro-inflammatory cytokines, such as IL-6, IL-1B, TNF-α, C-reactive protein. The cytokine expression shifts toward an IL-17 alloimmune responses with aging [75]. Adhesion molecules and chemokines play an important role in homing of lymphocytes and consequently in graft activation and rejection. They are generally increased by cellular stress [18]. It has been shown that T lymphocytes from elderly donors exhibited increased CD49d, CD50, and CD62L [76]. The vascular endothelium also regulates the postinflammatory fibroproliferative process [77]. Aging by itself enhances the sensibility of the endothelial cells to apoptotic stimuli and promotes morphological changes [78]. It has been shown that natural killer (NK) cells, key players of the innate immune system, have a decreased proliferative response upon stimulation with IL-2 [79].
Cells from the immune system are influenced by a variety of agents (hormones, cytokines, chemokines, adrenergic and cholinergic agonists, fatty acids, and immunoglobulins). The levels of many of these agents change with aging and can have a major impact on cell function. According to the theory proposed by Matzinger, the immune system recognizes any kind of cellular damage [80]. Thus, the aged microenvironment with increased cellular stress triggers “danger signals” and may facilitate activation of effector cells of the immune system instead of tolerogenic pathways. In addition, aged grafts are more susceptible to ischemia reperfusion injury because of increased production of reactive oxygen species, dysfunctional protective and repair mechanisms, leading to more damage and immune response [81, 82].
It has been shown that the aging process is associated with changes in the intracellular redox balance as a consequence of reduced capacity to produce active heat-shock proteins and reactive oxygen radicals scavengers [83, 84]. The healing process is also impaired with aging [85]. Cycles of injury and repair may have reached their limits, thus resulting in atrophy, uncontrolled proliferation, fibrosis, and impaired function [86, 87]. More recently, it has been shown that expression of Toll-like receptors, one of the receptors of the innate immune system, is altered in aged individuals [65, 74, 88, 89].
Aging and rejection
Both the age of donor and recipient play important role in allogenic response. In a large-multivariate analysis, donor age was identified as the most important risk factor for chronic kidney allograft failure [90]. The utilization of kidneys from old donors is also associated with an increased risk of delayed graft function (DGF) and acute rejection [91, 92]. It has been shown that donor age alone is representing a significant risk factor for patient death with a functioning kidney allograft, and it has been speculated that the poor function of the aged graft could lead to hypertension and an increased incidence of cardiovascular events [93]. Organs from elderly donors are particularly susceptible to ischemia/reperfusion injury, and associated with a higher incidence of DGF. For every 6 hours of cold ischemia, there is a 23% increase in the risk of DGF, thus increasing the risk for both, acute rejection and chronic graft deterioration [94].
Regarding the recipient age, it was commonly believed that older transplant recipients have a decreased risk to develop both acute and chronic rejection based on the concept of global deterioration of the immune system with aging [95]. To date, the reduced incidence of acute rejection has been confirmed in many experimental and clinical trials in cornea, kidney, cardiac, liver, and lung transplantation [92, 96]. The reduced incidence of acute rejection has been directly associated with an age-related immune dysfunction. Possible mechanisms to explain the decreased incidence of acute rejections in the elderly include: reduced numbers of naïve T cells, dysfunctional memory cells, increased sensibility to immunosuppression, reduced TCR-V and defective T-cell signaling, increased T-suppressor cells, and altered cytokine profiles.
Because of the reduced incidence of acute rejection in older recipients, allocation of older organs to older recipients may provide a better “immunological matching.” Graft survival was best among elderly recipients, after adjustment for death with a functioning graft. The frequency of acute rejection declined with every cohort of increasing recipient age. Donor age, in contrast, was associated with gradually increasing frequencies of rejection. This effect is blunted when old organs are transplanted into old recipients, and old recipients receiving old organs have excellent rates of graft and overall survival [92]. Thus, an “old-for old” program has been established in Europe—the Eurotransplant Senior Program [97], and proposed in the United States [98].
On the other hand, it has been shown that recipient age is also a strong and independent risk factor for the development of chronic allograft failure [99].
Important changes of the immune system that may increase the incidence of chronic rejection include increased numbers of memory T cells, low CD4+/CD8+ ratio, 31/32 shift [100], increased APC activation [19–21], upregulation of HLA-DR [101], production of antidonor HLA antibodies [42, 43], and increased pro-inflammatory cytokines (TNF-α, IL-4, IFN-γ, TGFβ-1, and IL-6) [99]. Age-related nonimmunological changes include impaired antioxidative and repair-remodeling abilities, and associated comorbidities (lipid disorders, diabetes, cardiovascular disease, hypertension, etc.). It has been shown that increased concentrations of homocysteine, apolipoproteins, and altered insulin-like growth factors accelerate the progression of arteriosclerosis in the elderly [99]. Age-related hormonal changes can also adversely affect the prognosis of transplantation in the elderly [19, 97, 102, 103].
Aging and tolerance
Billingham and Medawar long ago showed that age interferes with immunologic tolerance. They showed that a “window of opportunity” for immunologic tolerance exists for transplantation in neonates [104]. On the other hand, the aging immune system undergoes a series of changes that interfere with both the immune response of the recipient and the immunogenicity of the graft, which lead to reduced graft survival [5, 105].
The phenomenon of neonatal tolerance is multifactorial and there are many theories [106–110]. Based on findings from Burnet, Billingham, and Medawar, it has been theorized that experimental neonatal tolerance may occur by negative selection in the same way as natural self-tolerance develops [104, 106]. Another fact is that the neonate immune system is associated with decreased expression of MHC class II antigens and may not effectively presents antigens [109]. In addition, the neonatal immune system has little antigenic experience (naïve) and consequently reduced numbers of memory cells [111]. Memory cells are ready to mount a destructive response and constitute barriers to achieve graft tolerance. However, Ridge and Matzinger challenge this concept, proposing that tolerance is not an intrinsic property of the newborn immune system, but rather that the conditions under which the antigen is introduced determine whether the outcome is neonatal tolerance or immunization [110].
Aging is not only associated with increased resistance to achieve tolerance to foreign antigens [112] but also with loss of self-tolerance (autoimmunity). Aged animals and humans exhibit a decreased T-cell activation response although they have increased susceptibility to loss of self-tolerance. [38, 113–115].
Data on the effects of age on induction of tolerance to allografts is scarce. Most of the data on the effects of aging on immunologic tolerance come from animal experiments of oral tolerance to antigens. It has been shown in these studies that there is an age-dependent resistance to tolerance induction [116–124]. In transplantation, resistance to tolerance has been shown in a pancreas islet allograft model in the rat. Nine-month-old Lewis recipients were more resistant to intrathymic tolerance in comparison to 3-and 6-month-old recipients [41]. A study using anti-CD4 mAb (RIB 5/2) to induce tolerance to full-mismatched grafts of different age combinations in the rat showed that both the aged immune system and old graft interfere with tolerance and that these changes can be adoptively transferred to young recipients of young grafts [125]. Another study demonstrated that anti-CD45RB mAb therapy was ineffective in preventing rejection of cardiac grafts in aged recipients, although prolonged survival was similar to that observed in thymectomized young recipients [126]. In a large animal model for induction of tolerance to kidney allografts using cyclosporine, advanced age inhibited the establishment of tolerance [127]. Some studies showed that aged individuals may be rendered tolerant but may require that virtually all the potentially responsive cells be turned off and that the nature of antigen presentation sets the stage for immunization or tolerance [110, 116].
Changes in the innate and adaptive immune responses associated with aging may be obstacles to achieve immunologic tolerance. Potential mechanisms for this increased resistance are: (1) changes in frequencies and/or function of specific cellular subsets of the immune system deleting/anergizing; (2) different level of expression of adhesion, costimulatory, and MHC molecules, and heat-shock proteins; (3) the aged innate immune system/microenvironment (hormonal deficits, pro-oxidant and pro-inflammatory milieu), and (4) impaired ability of cellular repair.
MODULATION OF THE AGED IMMUNE SYSTEM
The increased resistance to achieve tolerance in aged individuals may be reversible. Modulation of the aged immune system by pharmacological or other means may represent a new avenue for prolongation of graft survival. Staples and colleagues showed successful induction of tolerance in older thymectomized and irradiated mice repopulated with lymphoid cells from young animals [118]. Other study showed that if aged mice resistant to tolerance induction are thymectomized, irradiated, and repopulated with spleen or bone marrow cells of young mice, they can become tolerant to specific antigens [118]. In another experiment, using a model of vascularized thymic transplantation in MHC inbred miniature swine it was shown that thymectomized recipients and aged recipients do not respond to an established protocol of tolerance induction with short-term tacrolimus. Surprisingly, aged thymic grafts transplanted into young recipients could be rejuvenated both histologically and functionally, suggesting that host environmental factors play an important role in thymic senescence. Interestingly, rejuvenated aged thymus could restore the ability to induce tolerance to kidney grafts across MHC class I mismatch [42]. Thymic function is reduced with aging but it is not lost, suggesting that therapeutic approaches to enhance thymic function may be successful even in very aged hosts [128].
Traditionally, approaches to improve the immune response in the elderly have focused on thymic rejuvenation [129–131]. It has been shown that many approaches can enhance thymopoieses including: administration of hormones and cytokines (growth hormone, prolactin, ghrelin, keratinocyte growth factor, vitamin D, and IL-7), androgen ablation, caloric restriction, bone marrow transplantation, and thymic tissue transplantation [131–140]. In the future, gene therapy and stem cell transplants may yield even better results.
A newer potential approach to enhance immunologic tolerance in the elderly is by modulation of tolerogenic cells. The potential of tolerogenic cells (regulatory T cells or tolerogenic DCs) transfer for the treatment of T-cell–mediated diseases (e.g. transplant rejection) in humans has gained momentum in recent years [141, 142].
It is unclear whether any single agent administered into older subjects can rejuvenate the immune system. However, combination of therapies to boost the aged immune system and reduce graft immunogenicity may improve outcomes.
CONCLUSIONS AND FUTURE PROSPECTS
Aging should be seen as a global and interconnected process of deterioration of all immune compartments. The aging process causes important anatomical and functional changes that result in reduction of the functional reserve and inability to cope with physiological and pathological stresses. In addition, changes associated with immunosenescence and the aging microenvironment may interfere with antigen recognition and efficient presentation, which in turn modifies both rejection and tolerance. The changes associated with immunosenescence are just beginning to be revealed.
Despite the aging population in most parts of the world, elderly patients continue to be underrepresented in transplant clinical trials [143]. To optimize the outcomes of transplantation in the elderly, it is necessary to test new protocols in the elderly population. Individualization of the immunosuppressive treatment and interventions to selectively target and reverse the crucial dysfunctions of the aging immune system and aged grafts are promising therapeutic strategies to improve graft and patient survival of the elderly transplant population.
Footnotes
Declaration of Interest
The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the article.
References
- 1.Department of Economic and Social Affairs Population Division. World population ageing 1950–2050. United Nations: 2002. pp. 15–22. [Google Scholar]
- 2. [Data accessed February 26th of 2013];OPTN home page. Available at: http://optn.transplant.hrsa.gov/data/
- 3.Pawelec G. Immunosenescence: impact in the young as well as the old? Mech Ageing Dev. 1999;108(1):1–7. doi: 10.1016/s0047-6374(99)00010-x. [DOI] [PubMed] [Google Scholar]
- 4.Castle SC. Clinical relevance of age-related immune dysfunction. Clin Infect Dis. 2000;31(2):578–585. doi: 10.1086/313947. [DOI] [PubMed] [Google Scholar]
- 5.Martins PN, Pratschke J, Pascher A, et al. Aging and immune response in organ transplantation. Transplantation. 2005;79(2):127–132. doi: 10.1097/01.tp.0000146258.79425.04. [DOI] [PubMed] [Google Scholar]
- 6.Epstein M. Aging and the kidney. J Am Soc Nephrol. 1996;7(8):1106–1122. doi: 10.1681/ASN.V781106. [DOI] [PubMed] [Google Scholar]
- 7.Halloran PF, Melk A, Barth C. Rethinking chronic allograft nephropathy: the concept of accelerated senescence. J Am Soc Nephrol. 1999;10(1):167–181. doi: 10.1681/ASN.V101167. [DOI] [PubMed] [Google Scholar]
- 8.Ginaldi L, De Martinis M, D’Ostilio A, et al. Immunological changes in the elderly. Aging. 1999;11(5):281–286. doi: 10.1007/BF03339801. [DOI] [PubMed] [Google Scholar]
- 9.Globerson A, Effros RB. Ageing of lymphocytes and lymphocytes in the aged. Immunol Today. 2000;21(10):515–521. doi: 10.1016/s0167-5699(00)01714-x. [DOI] [PubMed] [Google Scholar]
- 10.Geiger H, Van Zant G. The aging of lympho-hematopoietic stem cells. Nat Immunol. 2002;3(4):329–333. doi: 10.1038/ni0402-329. [DOI] [PubMed] [Google Scholar]
- 11.Beerman I, Maloney WJ, Weissmann IL, Rossi DJ. Stem cells and the aging hematopoietic system. Curr Opin Immunol. 2010a;22(4):500–506. doi: 10.1016/j.coi.2010.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cossarizza A, Ortolani C, Monti D, Francheschi C. Cytometric analysis of immunosenescence. Cytometry. 1997;27(4):297. doi: 10.1002/(sici)1097-0320(19970401)27:4<297::aid-cyto1>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 13.Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004;5(2):133–139. doi: 10.1038/ni1033. [DOI] [PubMed] [Google Scholar]
- 14.Lethonen L, Eskola J, Vanio O, Lethonen A. Changes in lymphocyte subsets and immune competence in very advanced age. J Gerontol. 1990;45(3):M108–112. doi: 10.1093/geronj/45.3.m108. [DOI] [PubMed] [Google Scholar]
- 15.Naylor K, Li G, Vallejo AN, et al. The influence of age on T cell generation and TCR diversity. J Immunol. 2005;174(11):7446–7452. doi: 10.4049/jimmunol.174.11.7446. [DOI] [PubMed] [Google Scholar]
- 16.Uyemura K, Castle SC, Makinodan T. The frail elderly: role of dendritic cells in the susceptibility to infection. Mech Ageing Dev. 2002;123(8):955–962. doi: 10.1016/s0047-6374(02)00033-7. [DOI] [PubMed] [Google Scholar]
- 17.Cossarizza A, Monti D, Cantini M, et al. Extremely low frequency pulsed electromagnetic fields increase interleukin-2 (IL-2) utilization and IL-2 receptor expression in lymphocytes from old subjects. FEBS Lett. 1989;248(1–2):141–144. doi: 10.1016/0014-5793(89)80449-1. [DOI] [PubMed] [Google Scholar]
- 18.Mo R, Chen J, Han Y, et al. T cell chemokine receptor expression in aging. J Immunol. 2003;170(2):895–904. doi: 10.4049/jimmunol.170.2.895. [DOI] [PubMed] [Google Scholar]
- 19.Carson MJ, Sutcliffe JG. Balancing function vs. self defense: the CNS as an active regulator of immune responses. J Neurosci Res. 1999;55(1):1–8. doi: 10.1002/(SICI)1097-4547(19990101)55:1<1::AID-JNR1>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
- 20.Lakkis FG, Sayegh MH. Memory T cells: a hurdle to immunologic tolerance. J Am Soc Nephrol. 2003;14(9):2402–2410. doi: 10.1097/01.asn.0000085020.78117.70. [DOI] [PubMed] [Google Scholar]
- 21.Croft M, Bradley LM, Swain SL. Naive versus memory CD4 T cell response to antigen; memory cells are less dependent on accessory cell costimulation and can respond to many APC types including resting B cells. J Immunol. 1994;152(6):2675–2685. [PubMed] [Google Scholar]
- 22.Dengler TJ, Pober JS. Human vascular endothelial cells stimulate memory but not naive CD8+ T cells to differentiate into CTL retaining an early activation phenotype. J Immunol. 2000;164(10):5146–5155. doi: 10.4049/jimmunol.164.10.5146. [DOI] [PubMed] [Google Scholar]
- 23.Bingaman AW, Farber DL. Memory T-cells in transplantation: generation, function, and potential role in rejection. Am J Transplant. 2004;4(6):846–852. doi: 10.1111/j.1600-6143.2004.00453.x. [DOI] [PubMed] [Google Scholar]
- 24.Inbrahim S, Dawson DW, Sanfilippo F. Predominant infiltration of rejecting human renal allografts with T cells expressing CD8 and CD45RO. Transplantation. 1995;59:724–728. doi: 10.1097/00007890-199503150-00015. [DOI] [PubMed] [Google Scholar]
- 25.Kline GH, Hayden TA, Klinman NR. B cell maintenance in aged mice reflects both increased B cell longevity and decreased B cell generation. J Immunol. 1999;162(6):3342–3349. [PubMed] [Google Scholar]
- 26.Gibson KL, Wu Y-C, Barnett Y, et al. B-cell diversity decreases in old age and is correlated with poor health status. Aging Cell. 2009;8(1):18–25. doi: 10.1111/j.1474-9726.2008.00443.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Colonna-Romano G, Bulati M, Aquino A, et al. B cells in the aged: CD27, CD5, and CD40 expression. Mech Ageing Dev. 2003;124(4):389–393. doi: 10.1016/s0047-6374(03)00013-7. [DOI] [PubMed] [Google Scholar]
- 28.Nossal GJ, Pike BL. Evidence for the clonal abortion theory of B-lymphocyte tolerance. J Exp Med. 1975;141(4):904–917. [PMC free article] [PubMed] [Google Scholar]
- 29.Cambier JC, Kettman JR, Vitetta ES, Uhr JW. Differential susceptibility of neonatal and adult murine spleen cells to in vitro induction of B-cell tolerance. J Exp Med. 1976;144(1):293–297. doi: 10.1084/jem.144.1.293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Metcalf ES, Klinman NR. In vitro tolerance induction of neonatal murine B cells. J Exp Med. 1976;143(6):1327–1340. doi: 10.1084/jem.143.6.1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Szewczuk MR, Siskind GW. Ontogeny of B-lymphocyte function. III. In vivo and in vitro studies on the ease of tolerance induction in B lymphocytes from fetal, neonatal, and adult mice. J Exp Med. 1977;145(6):1590–1601. doi: 10.1084/jem.145.6.1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.LeGuern C. Potential role of major histocompatibility complex class II peptides in regulatory tolerance to vascularized grafts. Transplantation. 2004;77(1 Suppl):S35–S37. doi: 10.1097/01.TP.0000106472.91343.8D. [DOI] [PubMed] [Google Scholar]
- 33.LeGuern C. Regulation of T-cell functions by MHC class II self-presentation. Trends Immunol. 2003;24(12):633–638. doi: 10.1016/j.it.2003.10.010. [DOI] [PubMed] [Google Scholar]
- 34.Benoist C, Mathis D. Regulation of major histocompatibility complex class-II genes: X, Y and other letters of the alphabet. Annu Rev Immunol. 1990;8:681–715. doi: 10.1146/annurev.iy.08.040190.003341. [DOI] [PubMed] [Google Scholar]
- 35.Janick-Buckner D, Briggs CJ, Meyer TE, et al. Major histocompatibility complex antigen expression on lymphocytes from aging strain A mice. Growth Dev Aging. 1991;55(1):53–62. [PubMed] [Google Scholar]
- 36.Pietschmann P, Hahn P, Kudlacek S, et al. Surface markers and transendothelial migration of dendritic cells from elderly subjects. Exp Gerontol. 2000;35(2):213–224. doi: 10.1016/s0531-5565(99)00089-3. [DOI] [PubMed] [Google Scholar]
- 37.Herrero C, Sebastián C, Marqués L, et al. Immunosenescence of macrophages: reduced MHC class II gene expression. Exp Gerontol. 2002;37(2–3):389–394. doi: 10.1016/s0531-5565(01)00205-4. [DOI] [PubMed] [Google Scholar]
- 38.Dekruyff RH, Kann EA, Weksler ME, Siskind GW. Effect of aging on T-cell tolerance induction. Cell Immunol. 1980;56(1):58–67. doi: 10.1016/0008-8749(80)90081-7. [DOI] [PubMed] [Google Scholar]
- 39.Dobken J, Weksler ME, Sikind GW. Effect of age on ease of B-cell tolerance induction. Cell Immunol. 1980;55(1):66–73. doi: 10.1016/0008-8749(80)90137-9. [DOI] [PubMed] [Google Scholar]
- 40.Dejaco C, Duftner C, Schirmer M. Are regulatory T-cells linked with aging? Exp Gerontol. 2006;41(4):339–345. doi: 10.1016/j.exger.2006.01.008. [DOI] [PubMed] [Google Scholar]
- 41.Qian T, Ricordi C, Inverardi L, Alejandro R. Intrathymic tolerance and age. Transplant Proc. 1995;27(6):3391. [PubMed] [Google Scholar]
- 42.Nobori S, Shimizu A, Okumi M, et al. Thymic rejuvenation and the induction of tolerance by adult thymic grafts. Proc Natl Acad Sci USA. 2006;103(50):19081–19086. doi: 10.1073/pnas.0605159103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhao L, Sun L, Wang H, et al. Changes of CD4+CD25+Foxp3+ regulatory T cells in aged Balb/c mice. J Leukoc Biol. 2007;81(6):1386–1394. doi: 10.1189/jlb.0506364. [DOI] [PubMed] [Google Scholar]
- 44.Sharma S, Dominguez AL, Lustgarten J. High accumulation of T regulatory cells prevents the activation of immune responses in aged animals. J Immunol. 2006;177(12):8348–5835. doi: 10.4049/jimmunol.177.12.8348. [DOI] [PubMed] [Google Scholar]
- 45.Trzonkowski P, Szmit E, Myśliwska J, Myśliwska A. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of CTL and NK cells in humans-impact of immunosenescence. Clin Immunol. 2006;119(3):307–316. doi: 10.1016/j.clim.2006.02.002. [DOI] [PubMed] [Google Scholar]
- 46.Hausman PB, Goidl EA, Siskind GW, Weksler ME. Immunological studies of aging. XI. Age-related changes in idiotype repertoire of suppressor T cells stimulated during tolerance induction. J Immunol. 1985;134(6):3802–3807. [PubMed] [Google Scholar]
- 47.Tsaknaridis L, Spencer L, Culbertson N, et al. Functional assay for human CD4+CD25+ Treg cells reveals an age-dependent loss of suppressive activity. J Neurosci Res. 2003;74(2):296–308. doi: 10.1002/jnr.10766. [DOI] [PubMed] [Google Scholar]
- 48.Kozlowska E, Biernacka M, Ciechomska M, Drela N. Age-related changes in the occurrence and characteristics of thymic CD4+ CD25+ T cells in mice. Immunology. 2007;122(3):445–453. doi: 10.1111/j.1365-2567.2007.02667.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Nishioka T, Shimizu J, Iida R, et al. CD4+CD25+Foxp3+ T cells and CD4+CD25-Foxp3+ T cells in aged mice. J Immunol. 2006;176(11):6586–6593. doi: 10.4049/jimmunol.176.11.6586. [DOI] [PubMed] [Google Scholar]
- 50.Wood KJ. Passenger leukocytes and microchimerism: what role in tolerance induction. Transplantation. 2003;75(9):17S–20S. doi: 10.1097/01.TP.0000067946.90241.2F. [DOI] [PubMed] [Google Scholar]
- 51.Effros R. Costimulatory pathway in the elderly. Vaccine. 2000;18(16):1661–1665. doi: 10.1016/s0264-410x(99)00503-4. [DOI] [PubMed] [Google Scholar]
- 52.Agrawal A, Agrawal S, Tay J, Gupta S. Biology of dendritic cells in aging. J Clin Immunol. 2008;28(1):14–20. doi: 10.1007/s10875-007-9127-6. [DOI] [PubMed] [Google Scholar]
- 53.Ordemann R, Hutchinson R, Friedman J, et al. Enhanced allostimulatory activity of host presenting cells in old mice intensifies acute graft-versus-host disease. J Clin Invest. 2002;109(9):1249–1256. doi: 10.1172/JCI14793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Castle CS, Uyemura K, Crawford W, et al. Antigen presenting cell function is enhanced in healthy elderly. Mech Ageing Dev. 1999;107(2):137–145. doi: 10.1016/s0047-6374(98)00141-9. [DOI] [PubMed] [Google Scholar]
- 55.Sidman CL, Luther EA, Marschall JD, et al. Increased expression of major histocompatibility complex antigens on lymphocytes from aged mice. Proc Natl Acad Sci USA. 1997;84(21):7624–7628. doi: 10.1073/pnas.84.21.7624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Della Bella S, Bierti L, Presicce P, et al. Peripheral blood dendritic cells and monocytes are differently regulated in the elderly. Clin Immunol. 2007;122(2):220–228. doi: 10.1016/j.clim.2006.09.012. [DOI] [PubMed] [Google Scholar]
- 57.Varas A, Sacedón R, Hernandez-López C, et al. Age-dependent changes in thymic macrophages and dendritic cells. Microsc Res Tech. 2003;62(6):501–507. doi: 10.1002/jemt.10411. [DOI] [PubMed] [Google Scholar]
- 58.Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
- 59.Leng Q, Bentwich Z, Borkow G. CTLA-4 upregulation during age. Mech Ageing Dev. 2002;123(10):1419–1421. doi: 10.1016/s0047-6374(02)00077-5. [DOI] [PubMed] [Google Scholar]
- 60.Azuma M, Phillips JH, Lanier LL. CD28-T lymphocytes: antigenic and functional properties. J Immunol. 1993;150(4):1147–1459. [PubMed] [Google Scholar]
- 61.Lio D, D’Anna C, Gervasi F, et al. Interleukin-12 release by mitogen-stimulated mononuclear cell in the elderly. Mech Ageing Dev. 1998;102(2–3):211–219. doi: 10.1016/s0047-6374(98)00016-5. [DOI] [PubMed] [Google Scholar]
- 62.Fernandez-Gutierrez B, Jover JA, De Miguel S, et al. Early lymphocyte activation in elderly humans: impaired T and T-dependent B cell responses. Exp Gerontol. 1999;34(2):217–229. doi: 10.1016/s0531-5565(98)00068-0. [DOI] [PubMed] [Google Scholar]
- 63.Fox A, Harrison LC. Innate immunity and graft rejection. Immunol Rev. 2000;173:141–147. doi: 10.1034/j.1600-065x.2000.917313.x. [DOI] [PubMed] [Google Scholar]
- 64.Palmer SM, Burch LH, Davis RD, et al. The role of innate immunity in acute allograft rejection after lung transplantation. Am J Respir Crit Care Med. 2003;168(6):628–632. doi: 10.1164/rccm.200303-447OC. [DOI] [PubMed] [Google Scholar]
- 65.Obhrai J, Goldstein DR. The role of toll-like receptors in solid organ transplantation. Transplantation. 2006;81(4):497–502. doi: 10.1097/01.tp.0000188124.42726.d8. [DOI] [PubMed] [Google Scholar]
- 66.Olszewski WL. Innate immunity processes in organ allografting—their contribution to acute and chronic rejection. Ann Transplant. 2005;10(2):5–9. [PubMed] [Google Scholar]
- 67.Gomez CR, Boehmer ED, Kovacs EJ. The aging innate immune system. Curr Opin Immunol. 2005;17(5):447–462. doi: 10.1016/j.coi.2005.07.013. [DOI] [PubMed] [Google Scholar]
- 68.LaRosa DF, Rahman AH, Turka LA. The innate immune system in allograft rejection and tolerance. J Immunol. 2007;178(12):7503–7509. doi: 10.4049/jimmunol.178.12.7503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Benichou G, Tonsho M, Tocco G, et al. Innate immunity and resistance to tolerogenesis in allotransplantation. Front Immunol. 2012;3:73. doi: 10.3389/fimmu.2012.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Plackett TP, Boehmer ED, Faunce DE, Kovacs EJ. Aging and innate immune cells. J Leukoc Biol. 2004;76(2):291–299. doi: 10.1189/jlb.1103592. [DOI] [PubMed] [Google Scholar]
- 71.Plowden J, Renshaw-Hoelscher M, Engleman C, et al. Innate immunity in aging: impact on macrophage function. Aging Cell. 2004;3(4):161–167. doi: 10.1111/j.1474-9728.2004.00102.x. [DOI] [PubMed] [Google Scholar]
- 72.Mishto M, Santoro A, Bellavista E, et al. Immunoproteasomes and immunosenescence. Ageing Res Rev. 2003;2(4):419–432. doi: 10.1016/s1568-1637(03)00030-8. [DOI] [PubMed] [Google Scholar]
- 73.Fulop T, Larbi A, Douziech N, et al. Signal transduction and functional changes in neutrophils with aging. Aging Cell. 2004;3(4):217–226. doi: 10.1111/j.1474-9728.2004.00110.x. [DOI] [PubMed] [Google Scholar]
- 74.Shaw AC, Panda A, Joshi SR, et al. Dysregulation of human Toll-like receptor function in aging. Ageing Res Rev. 2011;10(3):346–353. doi: 10.1016/j.arr.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tesar BM, Du W, Shirali AC, et al. Aging augments IL-17 T-cell alloimmune responses. Am J Transplant. 2009;9(1):54–63. doi: 10.1111/j.1600-6143.2008.02458.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.De Martinis M, Modesti M, Loreto MF, et al. Adhesion molecules on peripheral blood lymphocyte subpopulations in the elderly. Life Sci. 2000;68(2):139–151. doi: 10.1016/s0024-3205(00)00924-3. [DOI] [PubMed] [Google Scholar]
- 77.d’Alessio P. Aging and the endothelium. Exp Gerontol. 2004;39(2):165–171. doi: 10.1016/j.exger.2003.10.025. [DOI] [PubMed] [Google Scholar]
- 78.Hoffmann J, Haendeler J, Aicher A, et al. Aging enhances the sensitivity of endothelial cells toward apoptotic stimuli: important role of nitric oxide. Circ Res. 2001;89(8):709–715. doi: 10.1161/hh2001.097796. [DOI] [PubMed] [Google Scholar]
- 79.Borrego F, Alonso MC, Galiani MD, et al. NK phenotypic markers and IL2 response in NK cells from elderly people. Exp Gerontol. 1999;34(2):253–265. doi: 10.1016/s0531-5565(98)00076-x. [DOI] [PubMed] [Google Scholar]
- 80.Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol Rev. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
- 81.Park Y, Hirose R, Coatney JL, et al. Ischemia-reperfusion injury is more severe in older versus young rat livers. J Surg Res. 2007;137(1):96–102. doi: 10.1016/j.jss.2006.08.013. [DOI] [PubMed] [Google Scholar]
- 82.Okaya T, Blanchard J, Schuster R, et al. Age-dependent responses to hepatic ischemia/reperfusion injury. Shock. 2005;24(5):421–427. doi: 10.1097/01.shk.0000181282.14050.11. [DOI] [PubMed] [Google Scholar]
- 83.Verbeke P, Fonager J, Clark BF, Rattan SI. Heat shock response and aging: mechanisms and applications. Cell Biol Int. 2001;25(9):845–857. doi: 10.1006/cbir.2001.0789. [DOI] [PubMed] [Google Scholar]
- 84.Lee YK, Manalo D, Liu AK. Heat shock response, heat shock transcription factor and cell aging. Biol Signals. 1996;5(3):180–191. doi: 10.1159/000109187. [DOI] [PubMed] [Google Scholar]
- 85.Aschcroft GS, Mills SJ, Ashworth JJ. Ageing and wound healing. Biogerontology. 2002;3(6):337–345. doi: 10.1023/a:1021399228395. [DOI] [PubMed] [Google Scholar]
- 86.Safirstein RL, Bonventre JV. Molecular Nephrology: kidney function in health and disease. New York: Marcel Dekker; 1995. Molecular response to ischemic and nephrotoxic acute renal failure; p. 839. [Google Scholar]
- 87.Megyesi J, Udvarhelyi N, Safirstein RL, Price PM. The p53-independent activation of transcription of p21 WAF1/CIP1/SDI1 after acute renal failure. Am J Pathol. 1996;271(6):F1211–1216. doi: 10.1152/ajprenal.1996.271.6.F1211. [DOI] [PubMed] [Google Scholar]
- 88.Franceschi C, Bonafe M, Valensin S, et al. Inflammaging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–254. doi: 10.1111/j.1749-6632.2000.tb06651.x. [DOI] [PubMed] [Google Scholar]
- 89.Chelvarajan RL, Collins SM, Van Willigen JM, Bondada S. The unresponsiveness of aged mice to polysaccharide antigens is a result of a defect in macrophage function. J Leukoc Biol. 2005;77(4):503–512. doi: 10.1189/jlb.0804449. [DOI] [PubMed] [Google Scholar]
- 90.Gjertson DW. A multi-factor analysis of kidney graft outcomes at one and 5 years posttransplantation: 1996 UNOS update. Clin Transplant. 1996;343:360. [PubMed] [Google Scholar]
- 91.Terasaki PI, Gertson DW, Cecka JM, et al. Significance of the donor age effect on kidney transplants. Clin Transplant. 1997;11:366–372. [PubMed] [Google Scholar]
- 92.Tullius SG, Tran H, Guleria I, et al. The combination of donor and recipient age is critical in determining host immunoresponsiveness and renal transplant outcome. Ann Surg. 2010;252(4):662–674. doi: 10.1097/SLA.0b013e3181f65c7d. [DOI] [PubMed] [Google Scholar]
- 93.Meier-Kriesche HU, Cibrik DM, Ojo AO, et al. Interaction between donor and recipient age in determining the risk of chronic renal allograft failure. J Am Soc Nephrol. 2002;50(1):14–17. doi: 10.1046/j.1532-5415.2002.50002.x. [DOI] [PubMed] [Google Scholar]
- 94.Ojo AO, Wolfe RA, Held PJ, et al. Delayed graft function: risk factors and implications for renal allograft survival. Transplantation. 1997;63(7):968–974. doi: 10.1097/00007890-199704150-00011. [DOI] [PubMed] [Google Scholar]
- 95.Jassal SW, Olpez G, Cole E. Transplantation in the elderly: a review. Geriatr Nephrol Urol. 1997;7(3):157–165. doi: 10.1023/a:1008246129084. [DOI] [PubMed] [Google Scholar]
- 96.Brandley BA. Rejection and recipient age. Transpl Immunol. 2002;10(2–3):125–132. doi: 10.1016/s0966-3274(02)00058-8. [DOI] [PubMed] [Google Scholar]
- 97.Smits JM, Persijn GG, VanHouwelingen HC. Evaluation of the eurotransplant senior program. The results of the first year. Am J Transplant. 2002;2(7):664–670. doi: 10.1034/j.1600-6143.2002.20713.x. [DOI] [PubMed] [Google Scholar]
- 98.Leichtman AB, McCullough KP, Wolfe RA. Improving the allocation system for deceased-donor kidneys. N Engl J Med. 2011;364(14):1287–1289. doi: 10.1056/NEJMp1102728. [DOI] [PubMed] [Google Scholar]
- 99.Meier-Kriesche HU, Ojo AO, Cibrik DM, et al. Relationship of recipient age and the development of chronic allograft failure. Transplantation. 2000;70(2):306–310. doi: 10.1097/00007890-200007270-00012. [DOI] [PubMed] [Google Scholar]
- 100.Sandman M, Bruunsgaard H, Kemp K, et al. Is aging associated with a shift in the balance between type 1 and type 2 cytokines in humans? Clin Exp Immunol. 2002;127(1):107–114. doi: 10.1046/j.1365-2249.2002.01736.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rea IM, McNerlan SE, Alexander HD. CD69, CD25, and HLA-DR activation antigen expression on CD3+ lymphocytes and relationship to serum TNF-alpha, IFN-gamma, and sIL-2R levels in aging. Exp Gerontol. 1999;34(1):79–93. doi: 10.1016/s0531-5565(98)00058-8. [DOI] [PubMed] [Google Scholar]
- 102.Lou H, Kodama T, Zhao YJ, et al. Inhibition of transplant coronary arteriosclerosis in rabbits by chronic estradiol treatment is associated with abolition of MHC II antigen expression. Circulation. 1996;94(12):3355–3361. doi: 10.1161/01.cir.94.12.3355. [DOI] [PubMed] [Google Scholar]
- 103.Weidmann P, Demyttenaere-Bursztein S, Maxwell MH, DeLima J. Effect of aging on plasma renin and aldosterone in normal man. Kidney Int. 1975;8(5):325–333. doi: 10.1038/ki.1975.120. [DOI] [PubMed] [Google Scholar]
- 104.Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature. 1953;172(4279):603–606. doi: 10.1038/172603a0. [DOI] [PubMed] [Google Scholar]
- 105.Martins PN. Impact of donor and recipient age on allograft tolerance. Exp Clin Transplant. 2009;7(2):67–77. [PubMed] [Google Scholar]
- 106.Burnet FM. The clonal selection theory of acquired immunity. Nashville, TN: Vanderbilt University Press; 1959. [Google Scholar]
- 107.Vetro SW, Bellanti JA. Fetal and neonatal immunoincompetence. Fetal Ther. 1989;4(Suppl 1):82–91. doi: 10.1159/000263471. [DOI] [PubMed] [Google Scholar]
- 108.Streilein JW. Neonatal tolerance: towards an immunogenetic definition of self. Immunol Rev. 1979;46:123–146. doi: 10.1111/j.1600-065x.1979.tb00287.x. [DOI] [PubMed] [Google Scholar]
- 109.White DJ, Gilks W. The ontogeny of immune responses. J Heart Lung Transplant. 1993;12:S301–S308. [PubMed] [Google Scholar]
- 110.Ridge JP, Fuchs EJ, Matzinger P. Neonatal tolerance revisited: turning on newborn T-cells with dendritic cells. Science. 1996;271(5256):1723–1726. doi: 10.1126/science.271.5256.1723. [DOI] [PubMed] [Google Scholar]
- 111.Ruiz P, Nassiri M, Gregorian S, et al. Neonatal transplantation tolerance is associated with a systemic reduction in memory cells, altered chimeric cell phenotype, and modified ecosanoid and cytokine production. Transplantation. 1996;61(8):1198–1205. doi: 10.1097/00007890-199604270-00014. [DOI] [PubMed] [Google Scholar]
- 112.do Canto FB, Lima C, Junior, Teixeira IA, et al. Susceptibility of neonatal T cells and adult thymocytes to peripheral tolerance to allogeneic stimuli. Immunology. 2008;125(3):387–396. doi: 10.1111/j.1365-2567.2008.02855.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hsu HC, Scott DK, Mountz JD. Impaired apoptosis and immune senescence—cause or effect? Immunol Rev. 2005;205:130–146. doi: 10.1111/j.0105-2896.2005.00270.x. [DOI] [PubMed] [Google Scholar]
- 114.Prelog M. Aging of the immune system: a risk factor for autoimmunity? Autoimmun Rev. 2006;5(2):136–139. doi: 10.1016/j.autrev.2005.09.008. [DOI] [PubMed] [Google Scholar]
- 115.Urbán L, Bessenyei B, Márka M, Semsei I. On the role of aging in the etiology of autoimmunity. Gerontology. 2002;48(3):179–184. doi: 10.1159/000052839. [DOI] [PubMed] [Google Scholar]
- 116.Habicht GS. Acquired immunological tolerance in aged mice. I. The dose–response relationship. Mech Ageing Dev. 1982;19(1):53–62. doi: 10.1016/0047-6374(82)90050-1. [DOI] [PubMed] [Google Scholar]
- 117.Ponnappan U, Cinader B, Gerber V, Blaser K. Age-dependent changes in isotype expression and down-regulation of C57BL/6 mice. Scand J Immunol. 1987;25(1):45–54. doi: 10.1111/j.1365-3083.1987.tb01045.x. [DOI] [PubMed] [Google Scholar]
- 118.Staples PJ, Talal N. Relative inability to induce tolerance in adult NZB and NZB-NZWF1 mice. J Exp Med. 1969;129(1):123–139. doi: 10.1084/jem.129.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Amagai T, Nakano K, Cinader B. Mechanisms involved in age-dependent decline of immune responsiveness and apparent resistance against tolerance induction in C57BL/6 mice. Scand J Immunol. 1982;16(3):217–231. doi: 10.1111/j.1365-3083.1982.tb00717.x. [DOI] [PubMed] [Google Scholar]
- 120.de Faria AM, Ficker SM, Speziali E, et al. Aging affects oral tolerance induction but not its maintenance in mice. Mech Ageing Dev. 1998;102(1):67–80. doi: 10.1016/s0047-6374(98)00024-4. [DOI] [PubMed] [Google Scholar]
- 121.Wakabayashi A, Utsuyama M, Hosoda T, et al. Differential age effect of oral administration of an antigen on antibody response: an induction of tolerance in young mice but enhancement of immune response in old mice. Mech Ageing Dev. 1999;109(3):191–201. doi: 10.1016/s0047-6374(99)00036-6. [DOI] [PubMed] [Google Scholar]
- 122.Staples PJ, Steinberg AD, Talal N. Induction of immunologic tolerance in older New Zealand mice repopulated with young spleen, bone marrow, or thymus. J Exp Med. 1970;131(6):1223–1238. doi: 10.1084/jem.131.6.1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Vaz N, Faria MC, Verdolin A, Carvalho CR. Immaturity, ageing and oral tolerance. Scand J Immunol. 1997;46:225–229. doi: 10.1046/j.1365-3083.1997.d01-117.x. [DOI] [PubMed] [Google Scholar]
- 124.Gahring LC, Weigle WO. The effect of aging on the induction of humoral and cellular immunity and tolerance in two long-lived mouse strains. Cell Immunol. 1990;128(1):142–151. doi: 10.1016/0008-8749(90)90013-h. [DOI] [PubMed] [Google Scholar]
- 125.Martins PN, Reutzel-Selke A, Jurisch A, Tullius SG. “Donor and recipient age prevent tolerance induction” World Transplantation Congress (WTC) Am J Transplant. 2006;6(Suppl 2):895. [Google Scholar]
- 126.Huang X, Sonawane S, Kim J, et al. Age-dependent transplantation tolerance. Abstract 2513 WTC 2006. Am J Transplant. 2006;6(Suppl 2):890. [Google Scholar]
- 127.Yamada K, Gianello PR, Ierino FL, et al. Role of the thymus in transplantation tolerance in miniature swine: II. Effect of steroids and age on the induction of tolerance to class I mismatched renal allografts. Transplantation. 1999;67(3):458–467. doi: 10.1097/00007890-199902150-00020. [DOI] [PubMed] [Google Scholar]
- 128.Mackall CL, Gress RE. Thymic aging and T-cell regeneration. Immunol Rev. 1997;160:91–102. doi: 10.1111/j.1600-065x.1997.tb01030.x. [DOI] [PubMed] [Google Scholar]
- 129.Arlt W, Hewison M. Hormones and immune function: implications of aging. Aging Cell. 2004;3(4):209–216. doi: 10.1111/j.1474-9728.2004.00109.x. [DOI] [PubMed] [Google Scholar]
- 130.Taub DD, Longo DL. Insights into thymic aging and regeneration. Immunol Rev. 2005;205:72–93. doi: 10.1111/j.0105-2896.2005.00275.x. [DOI] [PubMed] [Google Scholar]
- 131.Fülöp T, Larbi A, Hirokawa K, et al. Immunosupportive therapies in aging. Clin Interv Aging. 2007;2(1):33–54. doi: 10.2147/ciia.2007.2.1.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Savino W, Postel-Vinay MC, Smaniotto S, Dardenne M. The thymus gland: a target organ for growth hormone. Scand J Immunol. 2002;55(5):442–452. doi: 10.1046/j.1365-3083.2002.01077.x. [DOI] [PubMed] [Google Scholar]
- 133.De Mello-Coelho V, Savino W, Postel-Vinay MC, Dardenne M. Role of prolactin and growth hormone on thymus physiology. Dev Immunol. 1998;6(3–4):317–323. doi: 10.1155/1998/89782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Dixit VD, Yang H, Sun Y, et al. Ghrelin promotes thymopoiesis during aging. J Clin Invest. 2007;117(10):2778–2790. doi: 10.1172/JCI30248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bruinsma M, van Soest PL, Leenen PJ, et al. Keratinocyte growth factor induces expansion of murine peripheral CD4+Foxp3+ regulatory T cells and increases their thymic output. J Immunol. 2007;179(11):7424–7430. doi: 10.4049/jimmunol.179.11.7424. [DOI] [PubMed] [Google Scholar]
- 136.Sportès C, Gress RE. Interleukin-7 immunotherapy. Adv Exp Med Biol. 2007;601:321–333. doi: 10.1007/978-0-387-72005-0_35. [DOI] [PubMed] [Google Scholar]
- 137.Henson SM, Pido-Lopez J, Aspinall R. Reversal of thymic atrophy. Exp Gerontol. 2004;39(4):673–678. doi: 10.1016/j.exger.2003.10.030. [DOI] [PubMed] [Google Scholar]
- 138.Wils EJ, Cornelissen JJ. Thymopoiesis following allogeneic stem cell transplantation: new possibilities for improvement. Blood Rev. 2005;19(2):89–98. doi: 10.1016/j.blre.2004.04.001. [DOI] [PubMed] [Google Scholar]
- 139.Haq AU. 1,25-Dihydroxyvitamin D3 (calcitriol) suppresses IL-2 induced murine thymocyte proliferation. Thymus. 1986;8(5):295–306. [PubMed] [Google Scholar]
- 140.Vallabhajosyula P, Griesemer A, Yamada K, et al. Vascularized composite islet-kidney transplantation in a miniature swine model. Cell Biochem Biophys. 2007;48(2–3):201–207. doi: 10.1007/s12013-007-0027-4. [DOI] [PubMed] [Google Scholar]
- 141.Bluestone JA. Regulatory T-cell therapy: is it ready for the clinic? Nat Rev Immunol. 2005;5(4):343–349. doi: 10.1038/nri1574. [DOI] [PubMed] [Google Scholar]
- 142.Bluestone JA, Thomson AW, Shevach EM, Weiner HL. What does the future hold for cell-based tolerogenic therapy? Nat Rev Immunol. 2007;7(8):650–654. doi: 10.1038/nri2137. [DOI] [PubMed] [Google Scholar]
- 143.Meyer JM, Archdeacon P, Albrecht R. FDA perspective: enrolment of elderly transplant recipients in clinical trials. Transplantation. 2013;95(7):916–918. doi: 10.1097/TP.0b013e31828279d9. [DOI] [PubMed] [Google Scholar]