Skip to main content
The Journals of Gerontology Series A: Biological Sciences and Medical Sciences logoLink to The Journals of Gerontology Series A: Biological Sciences and Medical Sciences
. 2012 Mar 5;67A(3):254–263. doi: 10.1093/gerona/glr237

T-Cell Biology in Aging, With a Focus on Lung Disease

Naeun Lee 1, Min Sun Shin 1, Insoo Kang 1,
PMCID: PMC3297764  PMID: 22396471

Abstract

T cells are essential for defending hosts against microorganisms and malignancy as well as for regulating the development of immune-mediated inflammatory diseases like autoimmunity. Alterations in T-cell immunity occur with aging, affecting the function and proportions of T-cell subsets. Probably, the most noticeable age-associated change in T-cell immunity is an alteration in the frequency of naive and memory CD4+ and CD8+ T cells. In fact, the frequency of naive CD4+ and CD8+ T cells decreases with aging, whereas the frequency of memory CD4+ and CD8+ T cells increases. Also, changes in T-cell proliferation, cytokine production, memory response, and cytotoxicity as well as in regulatory T-cell number and function have been reported with aging. Such alterations could contribute to the development of infections, malignancies, and inflammatory diseases that rise with aging. Of interest, T cells are closely involved in the development of inflammatory airway and lung diseases including asthma and chronic obstructive pulmonary disease, which are prevalent in the elderly people. In addition, T cells play a major role in defending host against influenza virus infection, a serious medical problem with high morbidity and mortality in the elderly people. Thus, it is conceivable that altered T-cell immunity may account in part for the development of such respiratory problems with aging. Here, we will review the recent advances in T-cell immunity and its alteration with aging and discuss the potential effects of such changes on the lung.

Keywords: T cells, Lungs, Aging


Alterations in the immune system occur with aging (1). These alterations likely contribute to the development of an array of clinical entities such as infections, malignancies, and inflammatory diseases that come across with aging. Although the increased frequency of infections and malignancies in the elderly people suggests an age-associated decline of the immune system (14), aging could be regarded as a condition with dysregulated inflammation (inflam-aging; (5,6)). The latter notion is supported by the observation that the elderly people had increased circulatory levels of the inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-α, which predicted mortality risk independently of other risk factors (7,8). Clinically, the elderly people have an increased incidence of some chronic inflammatory diseases including polymyalgia rheumatica, giant cell arteritis, and chronic obstructive pulmonary disease (COPD; (9,10)). In addition, atherosclerosis and type 2 diabetes, which are prevalent in the elderly people, are now considered chronic inflammatory conditions associated with an imbalance among proinflammatory and antiinflammatory cytokines (7,11).

Age-associated changes of the immune system are found in both innate and adaptive immunities (6,12). T cells, a component of the adaptive immunity, play a critical role in the immune system. T cells are essential for defending hosts against microorganisms and malignancy as well as for regulating the development of immune-mediated inflammatory diseases like autoimmunity. T cells are divided into CD4+ and CD8+ T cells based on the capacity to recognize the antigenic peptide presented in the context of the major histocompatibility complex class I and II molecules, respectively. CD4+ T cells provide help to other immune cells such as B cells and macrophages for their activation through producing cytokines and expressing costimulatory molecules (13,14). The primary function of CD8+ T cells armed with cytotoxic molecules is to kill host cells infected with pathogens or transformed into tumors (15). Indeed, CD4+ T cells and CD8+ T cells are referred to T helper (Th) and cytotoxic T cells, respectively. Here, we will review the latest findings on how aging affects T-cell immunity and discuss the potential effects of such changes on the lung.

OVERVIEW OF T-CELL BIOLOGY

Over the past two decades, our understanding of T-cell biology has substantially advanced. These include the identifications of new CD4+ T-cell subsets as well as the discovery of the mechanisms involved in memory T-cell homeostasis. Thus, it would be imperative to start the review by refreshing the general biology of T cells, focusing on the recent findings.

T-Cell Homeostasis

The development site of T cells is the thymus. The progenitor cells of T cells migrate from the bone marrow to the thymus where immature T cells or thymocytes undergo complicated selection processes to generate cells with the optimal affinity for the major histocompatibility complex molecules while avoiding the recognition of self-antigens (16). Cells that have survived the selection processes are exported into the circulation as naive but mature T cells. Naive T cells migrate to the secondary lymphoid tissues where they survey antigens presented by antigen-presenting cells, such as dendritic cells (14,15,17). When the naive T cells encounter the appropriate antigen, they become activated, proliferated, and differentiated into effector T cells, with the help from cognate interactions through costimulatory molecules and cytokines. The effector T cells move to the sites of infection and/or inflammation to control the source of antigens, such as pathogens. Upon the clearance of the source of the antigens, most effector T cells undergo activation-induced cell death, although a small number of cells survive and become memory T cells providing long-term immune protection against the same pathogens. The memory T cells can turn into effector cells, even in the absence of the help of costimulation and cytokines when they encounter the antigen again. At this time, the development speed and the quantity of this secondary immune response are much faster and greater than those of the primary immune response in the secondary lymphoid tissues.

CD4+ T-Cell Subsets: Th Cells and Regulatory T Cells

The concept of the existence of different CD4+ T-cell subsets with distinct functions was developed more than two decades ago. Indeed, CD4+ Th cells can be categorized into Th1, Th2, and Th17 cells with unique cellular characteristics based on the cytokines primarily produced by them. Although Th1 and Th2 cells predominantly produce interferon (IFN)-γ and IL-4, respectively, recently identified Th17 cells produce IL-17 (13,14,17,18). These cell subsets have distinctive roles in defending hosts against pathogens. Th1 cells are essential for controlling intracellular microorganisms such as mycobacteria by activating macrophages with IFN-γ. Parasites are effectively controlled by Th2 cells that produce the cytokines IL-5 and IL-13 with the capacity to activate eosinophils. Th17 cells are involved in eradicating extracellular microorganisms, including fungi, by activating neutrophils with IL-17. Although the proper immune responses by Th1, Th2, and Th17 cells are obviously beneficial for the host, any aberrant response can lead to the development of pathological conditions. For instance, allergy and asthma have been associated with increased Th2 cell response while dysregulated Th17 cell response is found in some autoimmune diseases, such as multiple sclerosis and systemic lupus erythematosus (14,19,20).

The differentiation of Th1, Th2, and Th17 cells from naive CD4+ T cells is critically dependent on cytokine milieu (13,14,17,18). IFN-γ and IL-12 induce Th1 cell differentiation, although IL-4 promotes Th2 cell development. In the differentiation of Th17 cells, multiple cytokines including IL-1β, IL-6, IL-21, IL-23, and transforming growth factor-β are known to enhance Th17 cell polarization. Along with the cytokines, transcription factors are also involved as the master regulators in the differentiation of Th cell subsets (13,14,17,18). The T-box transcription factor T-bet regulates Th1 cell differentiation. GATA3 and retinoid orphan receptor (ROR)γ t are essentially involved in the development of Th2 and Th17 cells, respectively. These transcription factors and cytokines operate reciprocally (13). The expression of GATA3 and RORγt can be suppressed by T-bet (21,22). Similarly, the differentiation of Th17 cells is repressed by the Th1 and Th2 cytokines IFN-γ and IL-4 (23).

In addition to Th cells, CD4+ T cells also have cell subsets with the immune regulatory function. In fact, the concept of suppressor T cells or regulatory T cells that could control immune responses was conceived in the early 1970s (24). However, it was nearly forgotten until the 1990s when several groups demonstrated the existence of T-cell subsets, which were able to regulate inflammatory immune responses in mice (2527). The best known one is naturally occurring CD4+ CD25+ regulatory T cells (Treg) that express the forkhead family transcriptional factor FOXP3 (28,29). The FOXP3 expression is essential for the regulatory of function. FOXP3 transfection to CD4+ CD25 T cells, which did not have regulatory function, conferred the immune regulatory property (3032). Also, mice and humans with mutations in the Foxp3 gene have immune-dysregulated phenotypes (28). Of interest, FOXP3 can be induced in CD4+ T cells without expressing FOXP3 by T-cell receptor (TCR) triggering, IL-2, and transforming growth factor-β (29). These cells also appear to have immune regulatory function and are called induced Treg cells as opposed to naturally occurring Treg cells that are developed in the thymus.

Effector CD8+ T Cells

Naive CD8+ T cells acquire cytotoxic function rapidly in the presence of antigenic stimulation and costimulation (33). This process is further enhanced by inflammatory cytokines including IL-12 and IFN-α (33). The acquisition of cytotoxic function with the expression of perforin and granzyme B is regulated by several transcription factors (34). The best known ones are T-bet and Eomesodermin that belong to the T-box transcription factor family. As discussed earlier, T-bet is the master transcription regulatory for Th1 cell differentiation. In CD8+ T cells, T-bet expression is upregulated in response to TCR, IFN-γ, and IL-12 stimulation (35,36). Similar to CD4+ T cells, T-bet is involved in IFN-γ production by CD8+ T cells, although it may not be critically required (37). Eomesodermin can also upregulate the cytotoxic molecules perforin and granzyme B as well as IFN-γ in CD8+ T cells (34). Although both T-bet and Eomesodermin are involved in inducing effector function in CD8+ T cells, they may have different roles in the development of memory CD8+ T cells. Although increased T-bet expression was associated with the generation of short-lived effector CD8+ T cells, the expression of Eomesodermin was required for the development of memory cells with the long-term survival capacity (35,38).

Memory T-Cell Subsets

Memory T cells are not a single population. Based on the capacity to migrate secondary lymphoid tissue and infected or inflamed peripheral sites, memory T cells can be categorized into central and effector memory (EM) T cells. Central memory T cells that express lymphoid tissue homing chemokine receptor 7 (CCR7) can migrate to secondary lymphoid tissues like the lymph nodes and spleen (39). In contrast, EM T cells can go to peripheral tissues such as the skin and mucosa through the expression of the receptors for the molecules expressed in inflamed tissues but not CCR7. In terms of functional capacity, naive and central memory cells produce IL-2 upon TCR triggering and have strong proliferative capacity. In comparison, EM cells can migrate to peripheral sites of inflammation via their expression of β1 and β2 integrins (40) as well as of receptors for inflammatory chemokines, such as CCR1, CCR3, and CCR5 (39,41). In addition, EM cells express cytotoxic molecules and produce effector cytokines, such as IFN-γ.

Development and Maintenance of Memory T Cells

Upon pathogen clearance, the majority of effector cells undergo activation-induced cell death, leaving behind a residual fraction of memory survivors that provide long-term protection against the same antigen. Memory T cells continue to divide at a slow rate in the absence of antigen, and IL-7 and IL-15 have emerged as the cytokines necessary for such homeostasis, as further discussed later (4244). IL-7 is largely produced by thymic epithelial and bone marrow stromal cells while the major source of IL-15 is cells of myeloid origin, including monocytes, macrophages, and dendritic cells (4446). IL-7 promoted survival of memory CD8+ T cells through upregulation of Bcl-2, an antiapoptotic molecule (42), whereas mice deficient of IL-15 or the IL-15 receptor had reduced numbers of memory CD8+ T cells with impaired cytotoxicity (43,44,47,48). In mice infected with virus, viral-specific CD8+ T cells that expressed high levels of the IL-7 receptor alpha chain (IL-7Rα) had the increased ability to become memory cells compared with the same cells expressing low levels of IL-7Rα (49). Similarly, human IL-7Rαhigh memory CD8+ T cells survived better in response to IL-7 compared with IL-7Rαlow memory CD8+ T cells (50). These findings suggest the role for IL-7 and its receptor in determining potential fates of effector CD8+ T cells. However, studies reported a redundant role for IL-7 and IL-7Rα in inducing and maintaining memory CD8+ T cells (5154). Mice deficient of IL-7 were still able to generate viral-specific CD8+ memory cells with increased Bcl-2 (52). IL-15 is likely accountable for this phenomenon (53). In the development of memory CD8+ T cells, transcription factors are also involved. T-bet and Blimp-1, encoded by the Prdm1 gene, were preferentially expressed in short-lived effector CD8+ T cells (35,55,56). In contrast, Eomesodermin expression was required for the development of memory cells with the long-term survival capacity (35,38). Analogous to CD8+ T cells, IL-7 is involved in the development and maintenance of memory CD4+ T cells (57,58). In addition, a recent study showed that IL-15 could maintain memory CD4+ T cells in mice with physiologically low levels of IL-7 (59). These findings indicate that IL-7 and IL-15 are involved in the maintenance of both memory CD4+ and CD8+ T cells.

AGING AND T-CELL IMMUNITY

Alterations in T-cell immunity occur with aging, affecting the function and proportions of T-cell subsets (Table 1; (12,6064)). In studying the effect of aging on T-cell function, it is important to consider the fact that aging affects the frequency of naive and memory T cells. The thymus, the development site of T cells, undergoes atrophy with aging (12). This has a direct impact on the proportions of naive and memory T cells. In aged animals and humans, the frequency of naive CD4+ and CD8+ T cells decreases, whereas the frequency of memory CD4+ and CD8+ T cells increases (12,6063). As discussed in the T-Cell Biology section, naive and memory T cells are clearly distinct populations with unique cellular characteristics. Thus, any age-associated change in T-cell function including proliferation and cytokine production could be secondary to the alteration in the frequency of naive and memory T cells.

Table 1.

Alterations in T-Cell Immunity With Aging

Thymus and T-Cell Subsets
    Thymus Atrophy
    Frequency of naive CD4+ and CD8+ T cells Decrease
    Frequency of memory CD4+ and CD8+ T cells Increase
CD4+ T cells
    Cell proliferation in response to PMA/ionomycin or PHA Decrease
    IFN-γ and IL-4 production from CD4+ T cells Variable (no change, decrease, and increase)
    Frequency of IL-17–producing Th17 cells in memory CD4+ T cell Decrease
    Frequency of FOXP3+ Treg cells No change to increase
CD8+ T cells
    Proliferation Decrease
    Cytotoxicity Decrease
    Frequency of IL-7Rαlow effector memory CD8+ T cells Increase
    Frequency of CD28− (antigen experienced) and CD57+ CD8+ T cells Increase
    T-cell receptor repertoires Decrease with oligoclonal CD8+ T-cell expansion

Note: PMA= phorbol myristate acetate; IFN = interferon; IL = interleukin; PHA = phytohemagglutinin.

Age-Associated Changes in CD4+ T Cells

Age-associated changes in the function and the frequency of CD4+ T-cell subsets are found in humans and mice (61,65,66). The elderly people have an increased frequency of memory CD4+ T cells and a decreased frequency of naive CD4+ T cells compared with the young (66). Age-associated alterations were also reported in CD4+ T-cell functions including TCR signaling, cognate helper function, vaccine response, cell proliferation, and cytokine production (66,67). However, the results of studies are not always uniform. In particular, some studies reported an age-associated decrease in CD4+ T-cell proliferation in response to strong T-cell stimulations, such as phytohemagglutinin, phorbol myristate acetate, and ionomycin (6870). In contrast, no altered proliferation of CD4+ T cells was reported when cells were stimulated with relatively low-dose anti-CD3 Abs (71,72). In consistence with these studies, we also noticed that young and elderly people had similar levels of CD4+ T-cell proliferation in response to low-dose anti-CD3 antibody stimulation, whereas the elderly people had decreased CD4+ T-cell proliferation in response to high-dose anti-CD3 antibody stimulation (73). Thus, it is likely that the proliferative capacity of CD4+ T cells from the young and the elderly people is comparable as these cells encounter relatively weak T-cell stimulation, even though such capacity is lower in the elderly people than in the young in response to strong and prolonged T-cell stimulation. A possible explanation for these findings is that the maximum proliferative capacity of CD4+ T cells in the elderly people is lower than that in the young, which likely stems from the shortening of a telomere length with aging (74,75).

Studies that investigated the effect of aging on Th1 and Th2 responses in humans and animals were largely done in the 1990s by measuring serum cytokine levels as well as analyzing the cytokine gene or protein production by total T cells or CD4+ T cells. The results of these studies were largely inconsistent. Although some studies reported an age-associated increase in IFN-γ production from T cells (7678), others reported no change (79,80) or even a decrease in the elderly people (8183). Similarly, the findings from studies comparing IL-4 production from T cells between the young and the elderly people are not consistent. In the elderly people, IL-4 levels were higher (84), lower (83), or similar (82) compared with the young. Recently, several studies investigated IL-17–producing Th17 cells in aging (85,86). The effect of aging on Th17 cells appears to be different between naive and memory CD4+ T cells in humans (86). In purified memory CD4+ T cells, the elderly people had a decreased frequency of Th17 cells compared with the young while both groups had similar frequencies of IFN-γ–producing Th1 cells. In contrast, the differentiation of IL-17–producing effector cells but not IFN-γ-producing cells from naive CD4+ T cells was enhanced in the elderly people compared with the young. The latter finding implies that the differentiation of Th17 cells from naive CD4+ T cells in response to new microorganism(s) may not be impaired in the elderly people despite the declined frequency of total naive CD4+ T cells. IL-1β is essential for the differentiation of Th17 cells. A recent study reported that naive CD4+ T cells with the expression of the IL-1 receptor 1 (IL-1R1) had increased differentiation of IL-17–producing CD4+ T cells from naive CD4+ T (87). In fact, IL-1R1 expression on naive CD4+ T cells was increased in the elderly people compared with the young, suggesting that such altered IL-1R1 expression on naive CD4+ T cells could be a contributing factor to the increased Th17 cell differentiation from naive CD4+ T cells in the elderly people.

The effect of aging on the number and function of Treg cells has been studied. The results of these studies are somewhat inconsistent (88). A moderately increased frequency of FOXP3+ CD4+ T cells or CD25+ CD4+ T cells with aging was reported in humans (8991). Also, a loss of the capacity to suppress target cell proliferation by CD4+ CD25+ T cells was found in the elderly people (92). In contrast, we found no difference in the frequency and phenotypic characteristics of FOXP3+ CD4+ T cells as well as their capacity to suppress inflammatory cytokine production and proliferation of CD4+ CD25− T cells between the young and the elderly people (93). However, the production of the antiinflammatory cytokine IL-10 from CD4+ CD25− T cells was more potently suppressed in the elderly people than in the young (93).

Age-Associated Changes in CD8+ T Cells

Age-associated alterations in CD8+ T cells have been extensively studied (2,94). These include impaired cellular functions like cytotoxicity as well as changes in the subsets of naive and memory CD8+ T cells. Aged hosts have a reduced ability to combat viral infections (9597), which correlate with impaired cytotoxicity (98,99) as well as impaired proliferation and IL-2 production by virus-specific memory CD8+ T cells (100,101). With aging, the frequency of naive CD8+ T cell decreases, whereas the frequency of memory CD8+ T cells increases (102104). In addition, oligoclonally expanded populations of CD8+ T cells appear in aged humans and mice (102,103,105), suggesting that the expansion of memory CD8+ T cells could be secondary to repeated exposures to microbial antigens over a lifetime. This notion is supported by the findings that expanded memory CD8+ T cells are typically CD28− (molecule downregulated on antigen-experienced cells), CCR7−, and CD57+ (replication senescence marker) with decreased length of the telomere, which becomes shortened with each cell replication (106,107). Furthermore, several studies reported an association between the infectious status of cytomegalovirus, which establishes life-long latent infection, and the increased prevalence of CD8+ T-cell oligoclonal expansion in elderly humans (62,108110). These observations suggest the potential role for chronic antigenic stimulations such as cytomegalovirus infection in expanding memory CD8+ T cells with aging (105,111,112). However, this may not be the only mechanism for such a phenomenon with aging. It is possible that the expansion of memory CD8+ T cells, including oligoclonally expanded cells, may occur independently of chronic antigenic stimulation via the alterations in IL-15- and/or IL-7-mediated CD8+ T-cell maintenance. This notion is supported by a study demonstrating the development of CD8+ T-cell clonal expansion in aged mice lacking a major histocompatibility complex class I molecule as well as in aged mice repeatedly injected with adjuvant alone, which stimulated many CD8+ T-cell clones nonspecifically (113). Of interest, my lab reported that human naive and central memory CD8+ T cells expressed high levels of IL-7Rα, whereas human EM CD8+ T cells (CCR7− CD45RA+/−) had two different subsets of cells expressing IL-7Rαhigh and IL-7Rαlow with distinct characteristics (50). The expression of the IL7Rα gene was differentially regulated in the two groups as determined by DNA methylation, an important gene regulatory mechanism that is inherited from mother to daughter cells in the IL-7Rα gene promoter (114). The elderly people had an expansion of IL-7Rαlow EM CD8+ T cells compared with the young. IL-7Rαlow EM CD8+ T cells were highly antigen-experienced cells with limited TCR repertoire and increased expression of the replication senescent marker CD57. These cells had impaired survival and replication in response to IL-7 and TCR triggering, respectively. However, IL-15 induced substantial proliferation of IL-7Rαlow cells in the presence and absence of TCR triggering, which supports the potential role for homeostatic cytokines such as IL-15 in expanding memory CD8+ T cells with aging (115). Although it is yet to be determined how the age-associated expansion of memory CD8+ T cells exactly affects the host immunity, such phenomenon could be harmful to hosts in that it may impair the ability of CD8+ T cells to properly develop immune responses to newly encountered microorganisms such as emerging strains of influenza virus by occupying “immunological space” (2,12,116118).

AGING, LUNG, AND T CELLS

The lung and immune system are closely coupled. The lung is constantly exposed to microorganisms and environmental irritants that can trigger the immune system. Any under or over immune responses to these stimuli could result in pathological conditions, such as pneumonia, asthma, and COPD. In fact, aging has been associated with an increased risk of influenza, pneumonia, and COPD (10,119). T cells are essential for the development of immune responses to influenza virus and vaccine (64). Studies reported age-associated alterations in T-cell immune responses to primary influenza virus infection and influenza vaccine. A decreased memory CD4+ T-cell response to the influenza vaccine had been reported in the elderly people (120). Also, the CD8+ T-cell response to the influenza virus diminishes with aging (121,122). A recent study reported that EM and effector CD8+ T cells in the elderly people had decreased cytotoxicity against the influenza virus compared with the young (121). In mice, TCR repertoire of CD8+ T cells responding to the influenza viral infection was restricted with aging, although the numbers of CD8+ T cells in the airways and lungs of young and aged mice were similar (122). Such impaired T-cell response to influenza virus could be related to the age-associated decrease in the frequency of naive T cells with the thymic atrophy (122). In a separate study, influenza virus–infected aged mice had increased mortality and delayed recovery compared with young mice infected with the same virus (123). This could be related to decreased lung infiltration of CD4+ and CD8+ T cells with reduced numbers of activated influenza viral–specific CD8+ T cells (123). Overall, these findings suggest that aging is associated with impaired influenza viral–specific T-cell responses that may stem from a decreased frequency of naive T cells as well as impaired function of memory and effector T cells.

T Cells in COPD

COPD that affects more than 2 million people worldwide is the fourth leading cause of death (124,125). The prevalence of COPD increases with aging independently of cigarette smoking, the best known risk factor for COPD (126,127). This suggests that age-associated change(s) could contribute to the development of COPD. A body of accumulating data indicates a critical role for the immune system in the pathogenesis of COPD (reviewed in (125)). Both innate and adaptive immune cells are likely involved in the development of dysregulated inflammation and tissue damage in COPD. T cells were frequently found in the airways of patients with COPD, suggesting the role of T-cell immunity in the pathogenesis of COPD (10,128). In fact, overexpression of the T-cell cytokines IFN-γ and IL-13 induced massive emphysematous changes in mice (129,130). CD8+ T cells with the cytotoxicity were the predominantly infiltrating T-cell type in the airways and alveolar compartment of patients with COPD (125,131). Also, the frequency of IFN-γ–producing CD8+ T cells in peripheral blood correlated with disease activity in patients with COPD (132). Deleting CD8+ T cells improved lung inflammation and tissue destruction in a mouse model of cigarette smoke–induced emphysema (133). These observations clearly support the critical implication of CD8+ T cells, which are armed with the capacity to produce cytotoxic molecules and inflammatory cytokines, in the pathogenesis of COPD.

Different types of CD4+ T cells appear to be associated with COPD. Numbers of CD4+ T cells, in particular IFN-γ–producing Th1 cells, were increased in the airways and parenchymal tissues of patients with COPD (134). In addition, increased expression of IL-17, a cytokine produced primarily by Th17 cells, was reported in bronchial mucosa and submucosa of patients with COPD (135,136). Furthermore, the increased number of IL-17–producing Th17 cells was found in the peripheral blood of patients with COPD (137). Of interest, the expansion of CD28− CD4+ T cells, which also increase with aging (138), was found in patients with COPD, correlating with impaired lung function (139). These cells had increased expression of natural killer–like T-cell receptors (CD94, CD158), intracellular perforin, and granzyme B (139). The results of studies measuring the frequency of Treg cells in bronchoalveolar lavage from patients with COPD were inconsistent, showing both increased and decreased numbers of this cell subset in patients with COPD (140,141). As discussed earlier, aging is associated with the altered numbers and functions of T cells including CD8+ T cells, Th17 cells, and Treg cells, which are likely involved in the pathogenesis of COPD. Thus, it is conceivable that such age-associated changes in T-cell immunity could contribute to the development and/or progress of COPD, which increases with aging. For instance, increased IL-17 production from naive CD4+ T cells in the elderly people could be an aggravating factor for COPD upon respiratory infection with a previously unexposed microorganism (86). Also, the expansion of EM CD8+ T cells with the expression of perforin in the elderly people could be detrimental to COPD (50) given the role of such cells in the pathogenesis of COPD. Further studies in basic and clinical science are warranted to address these intriguing questions, which will lead to better therapeutic interventions.

T Cells in Asthma

Although asthma has been considered a disease primarily affecting children and young adolescents, a substantial number of asthmatic patients develop the disease after the age of 40 (142). Furthermore, elderly asthmatic patients appear to have more severe disease (143). T cells are a major player in the pathogenesis of asthma (reviewed in (144)). Traditionally, asthma was thought to be a Th2 cell–mediated disease with the involvement of the cytokines IL-4, IL-5, and IL-13, which could promote IgE switching, eosinophilia, mast cell recruitment, mucus production, and airway hyperresponsiveness (144,145). However, recent studies indicate the involvement of other types of Th cells, Treg, and CD8+ T cells (144). Allergenic challenges promoted airway inflammation, hyperresponsiveness, and neutrophilia by inducing Th17-cell response (146,147). Th17 cells also enhanced Th2-mediated eosinophilic inflammation in mice (148). In addition, IL-9–producing CD4+ T helper cells (referred to Th9 cells) may participate in inducing airway inflammation and hyperresponsiveness, although its exact role is yet to be determined given the mixed results of the studies on IL-9 overexpression and IL-9 deficiency in a mouse model of asthma (149,150). Treg cells also appear to have a role in regulating airway inflammation in asthma. Transfer or induction of Treg cells reduced airway inflammation and airway hyperresonsiveness in mice and rats (151,152). CD8+ T cells are likely involved in the development of asthma. In the airways and sputum of patients with asthma, CD8+ T cells that produced IL-5, IL-13, and/or IFN-γ were found, correlating with disease activity (153155). In mice, deleting CD8+ T cells reduced allergic airway inflammation while reconstituting CD8+ T cells in these mice with EM but not central memory CD8+ T cells increased airway hyperresponsiveness, eosinophilic inflammation, and IL-13 levels in bronchoalveolar fluid (156). Although data regarding the effect of aging on the pathogenesis of asthma are limited, aging may affect airway hyperresponsiveness. In a mouse model of asthma, aged mice developed greater airway and lung inflammation in response to ovalbumin challenge compared with young mice (157). In addition, the cytokine profiles were different between the two groups. Aged mice had increased production of IL-5 and IFN-γ in the lung tissues and spleen cell culture, whereas young mice had increased levels of IL-4 and IL-13. These findings support the notion that aging may affect airway hyperresponsiveness and inflammation. Aging is associated with alterations in T-cell immunity, which is a critical component in the development of asthma. However, additional investigations are required to determine whether and how such alterations could promote asthma in humans.

In summary, a range of immunologic alterations in T-cell immunity occurs with aging, encompassing from cell function to the proportion of T-cell subsets. Despite extensive investigations, the mechanisms for and biological significance of such alterations are still largely unknown. Further studies are warranted to address these critical issues, in particular focusing on the organs that are frequently affected by aging (eg, lungs).

FUNDING

This work was supported in part by grants from the National Institutes of Health (AG028069 and AG030834 all to I.K.).

References

  • 1.Miller RA. Aging and immune function. In: Paul WE, editor. Fundamental Immunology. 4th ed. Philadelphia, PA: Lippincott-Raven; 1999. pp. 947–966. [Google Scholar]
  • 2.Effros RB, Cai Z, Linton PJ. CD8 T cells and aging. Crit Rev Immunol. 2003;23:45–64. doi: 10.1615/critrevimmunol.v23.i12.30. [DOI] [PubMed] [Google Scholar]
  • 3.Goronzy JJ, Weyand CM. T cell development and receptor diversity during aging. Curr Opin Immunol. 2005;17:468–475. doi: 10.1016/j.coi.2005.07.020. [DOI] [PubMed] [Google Scholar]
  • 4.Fulop T, Larbi A, Kotb R, de Angelis F, Pawelec G. Aging, immunity, and cancer. Discov Med. 2011;11:537–550. [PubMed] [Google Scholar]
  • 5.Cevenini E, Caruso C, Candore G, et al. Age-related inflammation: the contribution of different organs, tissues and systems. How to face it for therapeutic approaches. Curr Pharm Des. 2010;16:609–618. doi: 10.2174/138161210790883840. [DOI] [PubMed] [Google Scholar]
  • 6.Shaw AC, Joshi S, Greenwood H, Panda A, Lord JM. Aging of the innate immune system. Curr Opin Immunol. 2010;22:507–513. doi: 10.1016/j.coi.2010.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Huang CS, Chen SJ, Chung RL, Tang RB. Serum interleukin-5 measurements for monitoring acute asthma in children. J Asthma. 2005;42:297–300. doi: 10.1081/jas-200057886. [DOI] [PubMed] [Google Scholar]
  • 8.Krabbe KS, Pedersen M, Bruunsgaard H. Inflammatory mediators in the elderly. Exp Gerontol. 2004;39:687–699. doi: 10.1016/j.exger.2004.01.009. [DOI] [PubMed] [Google Scholar]
  • 9.Hunder GG. Clinical features of GCA/PMR. Clin Exp Rheumatol. 2000;18:S6–S8. [PubMed] [Google Scholar]
  • 10.Sharma G, Hanania NA, Shim YM. The aging immune system and its relationship to the development of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6:573–580. doi: 10.1513/pats.200904-022RM. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Vasto S, Candore G, Balistreri CR, et al. Inflammatory networks in ageing, age-related diseases and longevity. Mech Ageing Dev. 2007;128:83–91. doi: 10.1016/j.mad.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 12.Dorshkind K, Montecino-Rodriguez E, Signer RA. The ageing immune system: is it ever too old to become young again? Nat Rev Immunol. 2009;9:57–62. doi: 10.1038/nri2471. [DOI] [PubMed] [Google Scholar]
  • 13.Murphy KM, Stockinger B. Effector T cell plasticity: flexibility in the face of changing circumstances. Nat Immunol. 2010;11:674–680. doi: 10.1038/ni.1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reiner SL. Development in motion: helper T cells at work. Cell. 2007;129:33–36. doi: 10.1016/j.cell.2007.03.019. [DOI] [PubMed] [Google Scholar]
  • 15.Cui W, Kaech SM. Generation of effector CD8+ T cells and their conversion to memory T cells. Immunol Rev. 2010;236:151–166. doi: 10.1111/j.1600-065X.2010.00926.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Starr TK, Jameson SC, Hogquist KA. Positive and negative selection of T cells. Annu Rev Immunol. 2003;21:139–176. doi: 10.1146/annurev.immunol.21.120601.141107. [DOI] [PubMed] [Google Scholar]
  • 17.Zhu J, Paul WE. Heterogeneity and plasticity of T helper cells. Cell Res. 2010;20:4–12. doi: 10.1038/cr.2009.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dong C. TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat Rev Immunol. 2008;8:337–348. doi: 10.1038/nri2295. [DOI] [PubMed] [Google Scholar]
  • 19.Shah K, Lee WW, Lee SH, et al. Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus. Arthritis Res Ther. 2010;12:R53. doi: 10.1186/ar2964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. doi: 10.1146/annurev.immunol.021908.132710. [DOI] [PubMed] [Google Scholar]
  • 21.Usui T, Preiss JC, Kanno Y, et al. T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription. J Exp Med. 2006;203:755–766. doi: 10.1084/jem.20052165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lazarevic V, Chen X, Shim JH, et al. T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORgammat. Nat Immunol. 2011;12:96–104. doi: 10.1038/ni.1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Harrington LE. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:1123–1132. doi: 10.1038/ni1254. [DOI] [PubMed] [Google Scholar]
  • 24.Gershon RK, Cohen P, Hencin R, Liebhaber SA. Suppressor T cells. J Immunol. 1972;108:586–590. [PubMed] [Google Scholar]
  • 25.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 26.Chen Y, Kuchroo VK, Inobe J, Hafler DA, Weiner HL. Regulatory T cell clones induced by oral tolerance: suppression of autoimmune encephalomyelitis. Science. 1994;265:1237–1240. doi: 10.1126/science.7520605. [DOI] [PubMed] [Google Scholar]
  • 27.Groux H, O’Garra A, Bigler M, et al. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 1997;389:737–742. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
  • 28.Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. 2006;24:209–226. doi: 10.1146/annurev.immunol.24.021605.090547. [DOI] [PubMed] [Google Scholar]
  • 29.Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500. doi: 10.1038/nri2785. [DOI] [PubMed] [Google Scholar]
  • 30.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 31.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–1061. [PubMed] [Google Scholar]
  • 32.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–342. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
  • 33.Arens R, Schoenberger SP. Plasticity in programming of effector and memory CD8 T-cell formation. Immunol Rev. 2010;235:190–205. doi: 10.1111/j.0105-2896.2010.00899.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rutishauser RL, Kaech SM. Generating diversity: transcriptional regulation of effector and memory CD8 T-cell differentiation. Immunol Rev. 2010;235:219–233. doi: 10.1111/j.0105-2896.2010.00901.x. [DOI] [PubMed] [Google Scholar]
  • 35.Joshi NS, Cui W, Chandele A, et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity. 2007;27:281–295. doi: 10.1016/j.immuni.2007.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Takemoto N, Intlekofer AM, Northrup JT, Wherry EJ, Reiner SL. Cutting edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. J Immunol. 2006;177:7515–7519. doi: 10.4049/jimmunol.177.11.7515. [DOI] [PubMed] [Google Scholar]
  • 37.Szabo SJ, Sullivan BM, Stemmann C, Satoskar AR, Sleckman BP, Glimcher LH. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science. 2002;295:338–342. doi: 10.1126/science.1065543. [DOI] [PubMed] [Google Scholar]
  • 38.Rao RR, Li Q, Odunsi K, Shrikant PA. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity. 2010;32:67–78. doi: 10.1016/j.immuni.2009.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
  • 40.Baron JL, Madri JA, Ruddle NH, Hashim G, Janeway CA., Jr. Surface expression of alpha 4 integrin by CD4 T cells is required for their entry into brain parenchyma. J Exp Med. 1993;177:57–68. doi: 10.1084/jem.177.1.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tang HL, Cyster JG. Chemokine up-regulation and activated T cell attraction by maturing dendritic cells. Science. 1999;284:819–822. doi: 10.1126/science.284.5415.819. [DOI] [PubMed] [Google Scholar]
  • 42.Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance. J Immunol. 2005;174:6571–6576. doi: 10.4049/jimmunol.174.11.6571. [DOI] [PubMed] [Google Scholar]
  • 43.Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol. 2003;3:269–279. doi: 10.1038/nri1052. [DOI] [PubMed] [Google Scholar]
  • 44.Kim HR, Hwang KA, Park SH, Kang I. IL-7 and IL-15: biology and roles in T-Cell immunity in health and disease. Crit Rev Immunol. 2008;28:325–339. doi: 10.1615/critrevimmunol.v28.i4.40. [DOI] [PubMed] [Google Scholar]
  • 45.Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood. 2001;97:14–32. doi: 10.1182/blood.v97.1.14. [DOI] [PubMed] [Google Scholar]
  • 46.Miranda-Carus ME, Benito-Miguel M, Llamas MA, Balsa A, Martin-Mola E. Human T cells constitutively express IL-15 that promotes ex vivo T cell homeostatic proliferation through autocrine/juxtacrine loops. J Immunol. 2005;175:3656–3662. doi: 10.4049/jimmunol.175.6.3656. [DOI] [PubMed] [Google Scholar]
  • 47.Goldrath AW, Sivakumar PV, Glaccum M, et al. Cytokine requirements for acute and basal homeostatic proliferation of naive and memory CD8+ T cells. J Exp Med. 2002;195:1515–1522. doi: 10.1084/jem.20020033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Oh S, Perera LP, Burke DS, Waldmann TA, Berzofsky JA. IL-15/IL-15Ralpha-mediated avidity maturation of memory CD8+ T cells. Proc Natl Acad Sci U S A. 2004;101:15154–15159. doi: 10.1073/pnas.0406649101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kaech SM, Tan JT, Wherry EJ, Konieczny BT, Surh CD, Ahmed R. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat Immunol. 2003;4:1191–1198. doi: 10.1038/ni1009. [DOI] [PubMed] [Google Scholar]
  • 50.Kim HR, Hong MS, Dan JM, Kang I. Altered IL-7R{alpha} expression with aging and the potential implications of IL-7 therapy on CD8+ T-cell immune responses. Blood. 2006;107:2855–2862. doi: 10.1182/blood-2005-09-3560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tan JT, Ernst B, Kieper WC, LeRoy E, Sprent J, Surh CD. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J Exp Med. 2002;195:1523–1532. doi: 10.1084/jem.20020066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Klonowski KD, Williams KJ, Marzo AL, Lefrancois L. Cutting edge: IL-7-independent regulation of IL-7 receptor alpha expression and memory CD8 T cell development. J Immunol. 2006;177:4247–4251. doi: 10.4049/jimmunol.177.7.4247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Buentke E, Mathiot A, Tolaini M, Di Santo J, Zamoyska R, Seddon B. Do CD8 effector cells need IL-7R expression to become resting memory cells? Blood. 2006;108:1949–1956. doi: 10.1182/blood-2006-04-016857. [DOI] [PubMed] [Google Scholar]
  • 54.Hand TW, Morre M, Kaech SM. Expression of IL-7 receptor alpha is necessary but not sufficient for the formation of memory CD8 T cells during viral infection. Proc Natl Acad Sci U S A. 2007;104:11730–11735. doi: 10.1073/pnas.0705007104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kallies A, Xin A, Belz GT, Nutt SL. Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses. Immunity. 2009;31:283–295. doi: 10.1016/j.immuni.2009.06.021. [DOI] [PubMed] [Google Scholar]
  • 56.Rutishauser RL, Martins GA, Kalachikov S, et al. Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties. Immunity. 2009;31:296–308. doi: 10.1016/j.immuni.2009.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lenz DC, Kurz SK, Lemmens E, et al. IL-7 regulates basal homeostatic proliferation of antiviral CD4+T cell memory. Proc Natl Acad Sci U S A. 2004;101:9357–9362. doi: 10.1073/pnas.0400640101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Li J, Huston G, Swain SL. IL-7 promotes the transition of CD4 effectors to persistent memory cells. J Exp Med. 2003;198:1807–1815. doi: 10.1084/jem.20030725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Purton JF, Tan JT, Rubinstein MP, Kim DM, Sprent J, Surh CD. Antiviral CD4+ memory T cells are IL-15 dependent. J Exp Med. 2007;204:951–961. doi: 10.1084/jem.20061805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Nikolich-Zugich J. T cell aging: naive but not young. J Exp Med. 2005;201:837–840. doi: 10.1084/jem.20050341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Miller RA, Berger SB, Burke DT, et al. T cells in aging mice: genetic, developmental, and biochemical analyses. Immunol Rev. 2005;205:94–103. doi: 10.1111/j.0105-2896.2005.00254.x. [DOI] [PubMed] [Google Scholar]
  • 62.Akbar AN, Fletcher JM. Memory T cell homeostasis and senescence during aging. Curr Opin Immunol. 2005;17:480–485. doi: 10.1016/j.coi.2005.07.019. [DOI] [PubMed] [Google Scholar]
  • 63.Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004;5:133–139. doi: 10.1038/ni1033. [DOI] [PubMed] [Google Scholar]
  • 64.McElhaney JE, Effros RB. Immunosenescence: what does it mean to health outcomes in older adults? Curr Opin Immunol. 2009;21:418–424. doi: 10.1016/j.coi.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Maue AC, Haynes L. CD4+ T cells and immunosenescence—a mini-review. Gerontology. 2009;55:491–495. doi: 10.1159/000214842. [DOI] [PubMed] [Google Scholar]
  • 66.Kovaiou RD, Grubeck-Loebenstein B. Age-associated changes within CD4+ T cells. Immunol Lett. 2006;107:8–14. doi: 10.1016/j.imlet.2006.07.006. [DOI] [PubMed] [Google Scholar]
  • 67.Yung RL. Changes in immune function with age. Rheum Dis Clin North Am. 2000;26:455–473. doi: 10.1016/s0889-857x(05)70151-4. [DOI] [PubMed] [Google Scholar]
  • 68.Douziech N, Seres I, Larbi A, et al. Modulation of human lymphocyte proliferative response with aging. Exp Gerontol. 2002;37:369–387. doi: 10.1016/s0531-5565(01)00204-2. [DOI] [PubMed] [Google Scholar]
  • 69.Tortorella C, Pisconti A, Piazzolla G, Antonaci S. APC-dependent impairment of T cell proliferation in aging: role of CD28- and IL-12/IL-15-mediated signaling. Mech Ageing Dev. 2002;123:1389–1402. doi: 10.1016/s0047-6374(02)00079-9. [DOI] [PubMed] [Google Scholar]
  • 70.Schindowski K, Frohlich L, Maurer K, Muller WE, Eckert A. Age-related impairment of human T lymphocytes’ activation: specific differences between CD4(+) and CD8(+) subsets. Mech Ageing Dev. 2002;123:375–390. doi: 10.1016/s0047-6374(01)00396-7. [DOI] [PubMed] [Google Scholar]
  • 71.Beckman I, Dimopoulos K, Xu XN, Bradley J, Henschke P, Ahern M. T cell activation in the elderly: evidence for specific deficiencies in T cell/accessory cell interactions. Mech Ageing Dev. 1990;51:265–276. doi: 10.1016/0047-6374(90)90076-r. [DOI] [PubMed] [Google Scholar]
  • 72.Sansoni P, Fagnoni F, Vescovini R, et al. T lymphocyte proliferative capability to defined stimuli and costimulatory CD28 pathway is not impaired in healthy centenarians. Mech Ageing Dev. 1997;96:127–136. doi: 10.1016/s0047-6374(97)01887-3. [DOI] [PubMed] [Google Scholar]
  • 73.Hong MS, Dan JM, Lee WW, Kang I. Alterations in the kinetics of CD4+ T cells responses with aging. J Korean Rheum Assoc. 2009;16:271–280. [Google Scholar]
  • 74.Slagboom PE, Droog S, Boomsma DI. Genetic determination of telomere size in humans: a twin study of three age groups. Am J Hum Genet. 1994;55:876–882. [PMC free article] [PubMed] [Google Scholar]
  • 75.Weng NP, Hathcock KS, Hodes RJ. Regulation of telomere length and telomerase in T and B cells: a mechanism for maintaining replicative potential. Immunity. 1998;9:151–157. doi: 10.1016/s1074-7613(00)80597-x. [DOI] [PubMed] [Google Scholar]
  • 76.Chopra RK, Holbrook NJ, Powers DC, McCoy MT, Adler WH, Nagel JE. Interleukin 2, interleukin 2 receptor, and interferon-gamma synthesis and mRNA expression in phorbol myristate acetate and calcium ionophore A23187-stimulated T cells from elderly humans. Clin Immunol Immunopathol. 1989;53:297–308. doi: 10.1016/0090-1229(89)90058-5. [DOI] [PubMed] [Google Scholar]
  • 77.Sakata-Kaneko S, Wakatsuki Y, Matsunaga Y, Usui T, Kita T. Altered Th1/Th2 commitment in human CD4+ T cells with ageing. Clin Exp Immunol. 2000;120:267–273. doi: 10.1046/j.1365-2249.2000.01224.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yen CJ, Lin SL, Huang KT, Lin RH. Age-associated changes in interferon-gamma and interleukin-4 secretion by purified human CD4+ and CD8+ T cells. J Biomed Sci. 2000;7:317–321. doi: 10.1007/BF02253251. [DOI] [PubMed] [Google Scholar]
  • 79.Hessen MT, Kaye D, Murasko DM. Heterogeneous effects of exogenous lymphokines on lymphoproliferation of elderly subjects. Mech Ageing Dev. 1991;58:61–73. doi: 10.1016/0047-6374(91)90120-o. [DOI] [PubMed] [Google Scholar]
  • 80.Sindermann J, Kruse A, Frercks HJ, Schutz RM, Kirchner H. Investigations of the lymphokine system in elderly individuals. Mech Ageing Dev. 1993;70:149–159. doi: 10.1016/0047-6374(93)90066-z. [DOI] [PubMed] [Google Scholar]
  • 81.Abb J, Abb H, Deinhardt F. Age-related decline of human interferon alpha and interferon gamma production. Blut. 1984;48:285–289. doi: 10.1007/BF00320399. [DOI] [PubMed] [Google Scholar]
  • 82.Candore G, Di Lorenzo G, Melluso M, et al. gamma-Interferon, interleukin-4 and interleukin-6 in vitro production in old subjects. Autoimmunity. 1993;16:275–280. doi: 10.3109/08916939309014646. [DOI] [PubMed] [Google Scholar]
  • 83.Karanfilov CI, Liu B, Fox CC, Lakshmanan RR, Whisler RL. Age-related defects in Th1 and Th2 cytokine production by human T cells can be dissociated from altered frequencies of CD45RA+ and CD45RO+ T cell subsets. Mech Ageing Dev. 1999;109:97–112. doi: 10.1016/s0047-6374(99)00030-5. [DOI] [PubMed] [Google Scholar]
  • 84.Nijhuis EW, Remarque EJ, Hinloopen B, et al. Age-related increase in the fraction of CD27-CD4+ T cells and IL-4 production as a feature of CD4+ T cell differentiation in vivo. Clin Exp Immunol. 1994;96:528–534. doi: 10.1111/j.1365-2249.1994.tb06061.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Goetzl EJ, Huang MC, Kon J, et al. Gender specificity of altered human immune cytokine profiles in aging. FASEB J. 2010;24:3580–3589. doi: 10.1096/fj.10-160911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lee JS, Lee WW, Kim SH, et al. Age-associated alteration in naive and memory Th17 cell response in humans. Clin Immunol. 2011;140:84–91. doi: 10.1016/j.clim.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lee WW, Kang SW, Choi J, et al. Regulating human Th17 cells via differential expression of IL-1 receptor. Blood. 2010;115:530–540. doi: 10.1182/blood-2009-08-236521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang L, Xie Y, Zhu LJ, Chang TT, Mao YQ, Li J. An association between immunosenescence and CD4(+)CD25(+) regulatory T cells: a systematic review. Biomed Environ Sci. 2010;23:327–332. doi: 10.1016/S0895-3988(10)60072-4. [DOI] [PubMed] [Google Scholar]
  • 89.Lages CS, Suffia I, Velilla PA, et al. Functional regulatory T cells accumulate in aged hosts and promote chronic infectious disease reactivation. J Immunol. 2008;181:1835–1848. doi: 10.4049/jimmunol.181.3.1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gregg R, Smith CM, Clark FJ, et al. The number of human peripheral blood CD4+ CD25high regulatory T cells increases with age. Clin Exp Immunol. 2005;140:540–546. doi: 10.1111/j.1365-2249.2005.02798.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Trzonkowski P, Szmit E, Mysliwska J, Mysliwski A. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of CTL and NK cells in humans-impact of immunosenescence. Clin Immunol. 2006;119:307–316. doi: 10.1016/j.clim.2006.02.002. [DOI] [PubMed] [Google Scholar]
  • 92.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;74:296–308. doi: 10.1002/jnr.10766. [DOI] [PubMed] [Google Scholar]
  • 93.Hwang KA, Kim HR, Kang I. Aging and human CD4(+) regulatory T cells. Mech Ageing Dev. 2009;130:509–517. doi: 10.1016/j.mad.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Clambey ET, van Dyk LF, Kappler JW, Marrack P. Non-malignant clonal expansions of CD8+ memory T cells in aged individuals. Immunol Rev. 2005;205:170–189. doi: 10.1111/j.0105-2896.2005.00265.x. [DOI] [PubMed] [Google Scholar]
  • 95.Stein BE. Vaccinating elderly people. Protecting from avoidable disease. Drugs Aging. 1994;5:242–253. doi: 10.2165/00002512-199405040-00002. [DOI] [PubMed] [Google Scholar]
  • 96.Treanor J, Falsey A. Respiratory viral infections in the elderly. Antiviral Res. 1999;44:79–102. doi: 10.1016/S0166-3542(99)00062-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Webster RG. Immunity to influenza in the elderly. Vaccine. 2000;18:1686–1689. doi: 10.1016/s0264-410x(99)00507-1. [DOI] [PubMed] [Google Scholar]
  • 98.Kapasi ZF, Murali-Krishna K, McRae ML, Ahmed R. Defective generation but normal maintenance of memory T cells in old mice. Eur J Immunol. 2002;32:1567–1573. doi: 10.1002/1521-4141(200206)32:6<1567::AID-IMMU1567>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 99.Po JL, Gardner EM, Anaraki F, Katsikis PD, Murasko DM. Age-associated decrease in virus-specific CD8+ T lymphocytes during primary influenza infection. Mech Ageing Dev. 2002;123:1167–1181. doi: 10.1016/s0047-6374(02)00010-6. [DOI] [PubMed] [Google Scholar]
  • 100.Effros RB, Walford RL. The immune response of aged mice to influenza: diminished T-cell proliferation, interleukin 2 production and cytotoxicity. Cell Immunol. 1983;81:298–305. doi: 10.1016/0008-8749(83)90237-x. [DOI] [PubMed] [Google Scholar]
  • 101.Effros RB, Walford RL. Diminished T-cell response to influenza virus in aged mice. Immunology. 1983;49:387–392. [PMC free article] [PubMed] [Google Scholar]
  • 102.Callahan JE, Kappler JW, Marrack P. Unexpected expansions of CD8-bearing cells in old mice. J Immunol. 1993;151:6657–6669. [PubMed] [Google Scholar]
  • 103.Posnett DN, Sinha R, Kabak S, Russo C. Clonal populations of T cells in normal elderly humans: the T cell equivalent to “benign monoclonal gammapathy”. doi: 10.1084/jem.179.2.609. [published erratum appears in J Exp Med 1994 Mar 1;179(3):1077]. J Exp Med. 1994;179:609–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hong MS, Dan JM, Choi JY, Kang I. Age-associated changes in the frequency of naive, memory and effector CD8+ T cells. Mech Ageing Dev. 2004;125:615–618. doi: 10.1016/j.mad.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 105.Clambey ET, Kappler JW, Marrack P. CD8 T cell clonal expansions & aging: a heterogeneous phenomenon with a common outcome. Exp Gerontol. 2007;42:407–411. doi: 10.1016/j.exger.2006.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Batliwalla FM, Rufer N, Lansdorp PM, Gregersen PK. Oligoclonal expansions in the CD8(+)CD28(-) T cells largely explain the shorter telomeres detected in this subset: analysis by flow FISH. Hum Immunol. 2000;61:951–958. doi: 10.1016/s0198-8859(00)00157-9. [DOI] [PubMed] [Google Scholar]
  • 107.Monteiro J, Batliwalla F, Ostrer H, Gregersen PK. Shortened telomeres in clonally expanded CD28-CD8+ T cells imply a replicative history that is distinct from their CD28+CD8+ counterparts. J Immunol. 1996;156:3587–3590. [PubMed] [Google Scholar]
  • 108.Khan N, Shariff N, Cobbold M, et al. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J Immunol. 2002;169:1984–1992. doi: 10.4049/jimmunol.169.4.1984. [DOI] [PubMed] [Google Scholar]
  • 109.Ouyang Q, Wagner WM, Voehringer D, et al. Age-associated accumulation of CMV-specific CD8+ T cells expressing the inhibitory killer cell lectin-like receptor G1 (KLRG1) Exp Gerontol. 2003;38:911–920. doi: 10.1016/s0531-5565(03)00134-7. [DOI] [PubMed] [Google Scholar]
  • 110.Almanzar G, Schwaiger S, Jenewein B, et al. Long-term cytomegalovirus infection leads to significant changes in the composition of the CD8+ T-cell repertoire, which may be the basis for an imbalance in the cytokine production profile in elderly persons. J Virol. 2005;79:3675–3683. doi: 10.1128/JVI.79.6.3675-3683.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Vasto S, Colonna-Romano G, Larbi A, Wikby A, Caruso C, Pawelec G. Role of persistent CMV infection in configuring T cell immunity in the elderly. Immun Ageing. 2007;4:2. doi: 10.1186/1742-4933-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Koch S, Larbi A, Ozcelik D, et al. Cytomegalovirus infection: a driving force in human T cell immunosenescence. Ann N Y Acad Sci. 2007;1114:23–35. doi: 10.1196/annals.1396.043. [DOI] [PubMed] [Google Scholar]
  • 113.Messaoudi I, Warner J, Nikolich-Zugich D, Fischer M, Nikolich-Zugich J. Molecular, cellular, and antigen requirements for development of age-associated T cell clonal expansions in vivo. J Immunol. 2006;176:301–308. doi: 10.4049/jimmunol.176.1.301. [DOI] [PubMed] [Google Scholar]
  • 114.Kim HR, Hwang KA, Kim KC, Kang I. Down-regulation of IL-7Ralpha expression in human T cells via DNA methylation. J Immunol. 2007;178:5473–5479. doi: 10.4049/jimmunol.178.9.5473. [DOI] [PubMed] [Google Scholar]
  • 115.Kim HR, Hwang KA, Kang I. Dual roles of IL-15 in maintaining IL-7RalphalowCCR7 memory CD8+ T cells in humans via recovering the phosphatidylinositol 3-kinase/AKT pathway. J Immunol. 2007;179:6734–6740. doi: 10.4049/jimmunol.179.10.6734. [DOI] [PubMed] [Google Scholar]
  • 116.Nikolich-Zugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat Rev Immunol. 2008;8:512–522. doi: 10.1038/nri2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Messaoudi I, LeMaoult J, Guevara-Patino JA, Metzner BM, Nikolich-Zugich J. Age-related CD8 T cell clonal expansions constrict CD8 T cell repertoire and have the potential to impair immune defense. J Exp Med. 2004;200:1347–1358. doi: 10.1084/jem.20040437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Saurwein-Teissl M, Lung TL, Marx F, 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]
  • 119.Meyer KC. The role of immunity and inflammation in lung senescence and susceptibility to infection in the elderly. Semin Respir Crit Care Med. 2010;31:561–574. doi: 10.1055/s-0030-1265897. [DOI] [PubMed] [Google Scholar]
  • 120.Kang I, Hong MS, Nolasco H, et al. Age-associated change in the frequency of memory CD4+ T cells impairs long term CD4+ T cell responses to influenza vaccine. J Immunol. 2004;173:673–681. doi: 10.4049/jimmunol.173.1.673. [DOI] [PubMed] [Google Scholar]
  • 121.Zhou X, McElhaney JE. Age-related changes in memory and effector T cells responding to influenza A/H3N2 and pandemic A/H1N1 strains in humans. Vaccine. 2011;29:2169–2177. doi: 10.1016/j.vaccine.2010.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yager EJ, Ahmed M, Lanzer K, Randall TD, Woodland DL, Blackman MA. Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus. J Exp Med. 2008;205:711–723. doi: 10.1084/jem.20071140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Toapanta FR, Ross TM. Impaired immune responses in the lungs of aged mice following influenza infection. Respir Res. 2009;10:112. doi: 10.1186/1465-9921-10-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ. Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006;367:1747–1757. doi: 10.1016/S0140-6736(06)68770-9. [DOI] [PubMed] [Google Scholar]
  • 125.Brusselle GG, Joos GF, Bracke KR. New insights into the immunology of chronic obstructive pulmonary disease. Lancet. 2011;378:1015–1026. doi: 10.1016/S0140-6736(11)60988-4. [DOI] [PubMed] [Google Scholar]
  • 126.Behrendt CE. Mild and moderate-to-severe COPD in nonsmokers: distinct demographic profiles. Chest. 2005;128:1239–1244. doi: 10.1378/chest.128.3.1239. [DOI] [PubMed] [Google Scholar]
  • 127.Buist AS, McBurnie MA, Vollmer WM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007;370:741–750. doi: 10.1016/S0140-6736(07)61377-4. [DOI] [PubMed] [Google Scholar]
  • 128.Gorska K, Maskey-Warzechowska M, Krenke R. Airway inflammation in chronic obstructive pulmonary disease. Curr Opin Pulm Med. 2010;16:89–96. doi: 10.1097/MCP.0b013e3283341ba0. [DOI] [PubMed] [Google Scholar]
  • 129.Zheng T, Zhu Z, Wang Z, et al. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest. 2000;106:1081–1093. doi: 10.1172/JCI10458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wang Z, Zheng T, Zhu Z, et al. Interferon gamma induction of pulmonary emphysema in the adult murine lung. J Exp Med. 2000;192:1587–1600. doi: 10.1084/jem.192.11.1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360:2445–2454. doi: 10.1056/NEJMra0804752. [DOI] [PubMed] [Google Scholar]
  • 132.Gadgil A, Zhu X, Sciurba FC, Duncan SR. Altered T-cell phenotypes in chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2006;3:487–488. doi: 10.1513/pats.200603-064MS. [DOI] [PubMed] [Google Scholar]
  • 133.Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA, Shapiro SD. CD8+ T cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice. J Immunol. 2007;178:8090–8096. doi: 10.4049/jimmunol.178.12.8090. [DOI] [PubMed] [Google Scholar]
  • 134.Grumelli S, Corry DB, Song LZ, et al. An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLoS Med. 2004;1:e8. doi: 10.1371/journal.pmed.0010008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Di Stefano A, Caramori G, Gnemmi I, et al. T helper type 17-related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin Exp Immunol. 2009;157:316–324. doi: 10.1111/j.1365-2249.2009.03965.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Doe C, Bafadhel M, Siddiqui S, et al. Expression of the T helper 17-associated cytokines IL-17A and IL-17F in asthma and COPD. Chest. 2010;138:1140–1147. doi: 10.1378/chest.09-3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Vargas-Rojas MI, Ramirez-Venegas A, Limon-Camacho L, Ochoa L, Hernandez-Zenteno R, Sansores RH. Increase of Th17 cells in peripheral blood of patients with chronic obstructive pulmonary disease. Respir Med. 2011;105:1648–1654. doi: 10.1016/j.rmed.2011.05.017. [DOI] [PubMed] [Google Scholar]
  • 138.Weyand CM, Brandes JC, Schmidt D, Fulbright JW, Goronzy JJ. Functional properties of CD4+ CD28- T cells in the aging immune system. Mech Ageing Dev. 1998;102:131–147. doi: 10.1016/s0047-6374(97)00161-9. [DOI] [PubMed] [Google Scholar]
  • 139.Lambers C, Hacker S, Posch M, et al. T cell senescence and contraction of T cell repertoire diversity in patients with chronic obstructive pulmonary disease. Clin Exp Immunol. 2009;155:466–475. doi: 10.1111/j.1365-2249.2008.03835.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Barcelo B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agusti AG. Phenotypic characterisation of T-lymphocytes in COPD: abnormal CD4+CD25+ regulatory T-lymphocyte response to tobacco smoking. Eur Respir J. 2008;31:555–562. doi: 10.1183/09031936.00010407. [DOI] [PubMed] [Google Scholar]
  • 141.Smyth LJ, Starkey C, Vestbo J, Singh D. CD4-regulatory cells in COPD patients. Chest. 2007;132:156–163. doi: 10.1378/chest.07-0083. [DOI] [PubMed] [Google Scholar]
  • 142.Broder I, Higgins MW, Mathews KP, Keller JB. Epidemiology of asthma and allergic rhinitis in a total community, Tecumseh, Michigan. 3. Second survey of the community. J Allergy Clin Immunol. 1974;53:127–138. doi: 10.1016/0091-6749(74)90001-3. [DOI] [PubMed] [Google Scholar]
  • 143.Miranda C, Busacker A, Balzar S, Trudeau J, Wenzel SE. Distinguishing severe asthma phenotypes: role of age at onset and eosinophilic inflammation. J Allergy Clin Immunol. 2004;113:101–108. doi: 10.1016/j.jaci.2003.10.041. [DOI] [PubMed] [Google Scholar]
  • 144.Lloyd CM, Hessel EM. Functions of T cells in asthma: more than just T(H)2 cells. Nat Rev Immunol. 2010;10:838–848. doi: 10.1038/nri2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Cohn L, Elias JA, Chupp GL. Asthma: mechanisms of disease persistence and progression. Annu Rev Immunol. 2004;22:789–815. doi: 10.1146/annurev.immunol.22.012703.104716. [DOI] [PubMed] [Google Scholar]
  • 146.He R, Oyoshi MK, Jin H, Geha RS. Epicutaneous antigen exposure induces a Th17 response that drives airway inflammation after inhalation challenge. Proc Natl Acad Sci U S A. 2007;104:15817–15822. doi: 10.1073/pnas.0706942104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wilson RH, Whitehead GS, Nakano H, Free ME, Kolls JK, Cook DN. Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness. Am J Respir Crit Care Med. 2009;180:720–730. doi: 10.1164/rccm.200904-0573OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wakashin H, Hirose K, Maezawa Y, et al. IL-23 and Th17 cells enhance Th2-cell-mediated eosinophilic airway inflammation in mice. Am J Respir Crit Care Med. 2008;178:1023–1032. doi: 10.1164/rccm.200801-086OC. [DOI] [PubMed] [Google Scholar]
  • 149.Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of interleukin 9 in the lungs of transgenic mice causes airway inflammation, mast cell hyperplasia, and bronchial hyperresponsiveness. J Exp Med. 1998;188:1307–1320. doi: 10.1084/jem.188.7.1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.McMillan SJ, Bishop B, Townsend MJ, McKenzie AN, Lloyd CM. The absence of interleukin 9 does not affect the development of allergen-induced pulmonary inflammation nor airway hyperreactivity. J Exp Med. 2002;195:51–57. doi: 10.1084/jem.20011732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Kearley J, Barker JE, Robinson DS, Lloyd CM. Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4+CD25+ regulatory T cells is interleukin 10 dependent. J Exp Med. 2005;202:1539–1547. doi: 10.1084/jem.20051166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Strickland DH, Stumbles PA, Zosky GR, et al. Reversal of airway hyperresponsiveness by induction of airway mucosal CD4+CD25+ regulatory T cells. J Exp Med. 2006;203:2649–2660. doi: 10.1084/jem.20060155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Cho SH, Stanciu LA, Holgate ST, Johnston SL. Increased interleukin-4, interleukin-5, and interferon-gamma in airway CD4+ and CD8+ T cells in atopic asthma. Am J Respir Crit Care Med. 2005;171:224–230. doi: 10.1164/rccm.200310-1416OC. [DOI] [PubMed] [Google Scholar]
  • 154.Ying S, Humbert M, Barkans J, et al. Expression of IL-4 and IL-5 mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol. 1997;158:3539–3544. [PubMed] [Google Scholar]
  • 155.van Rensen EL, Sont JK, Evertse CE, et al. Bronchial CD8 cell infiltrate and lung function decline in asthma. Am J Respir Crit Care Med. 2005;172:837–841. doi: 10.1164/rccm.200504-619OC. [DOI] [PubMed] [Google Scholar]
  • 156.Miyahara N, Swanson BJ, Takeda K, et al. Effector CD8+ T cells mediate inflammation and airway hyper-responsiveness. Nat Med. 2004;10:865–869. doi: 10.1038/nm1081. [DOI] [PubMed] [Google Scholar]
  • 157.Busse PJ, Zhang TF, Srivastava K, Schofield B, Li XM. Effect of ageing on pulmonary inflammation, airway hyperresponsiveness and T and B cell responses in antigen-sensitized and -challenged mice. Clin Exp Allergy. 2007;37:1392–1403. doi: 10.1111/j.1365-2222.2007.02775.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journals of Gerontology Series A: Biological Sciences and Medical Sciences are provided here courtesy of Oxford University Press

RESOURCES