Summary
A detailed understanding of the immune response to human immunodeficiency virus (HIV) infection is needed to inform prevention and therapeutic strategies that aim to contain the acquired immunodeficiency syndrome (AIDS) pandemic. The cellular immune response plays a critical role in controlling viral replication during HIV infection and will likely need to be a part of any vaccine approach. The qualitative feature of the cellular response most closely associated with immunologic control of HIV infection is CD8+ T-cell cytotoxic potential, which is responsible for mediating the elimination of infected CD4+ T cells. Understanding the underlying mechanisms involved in regulating the elicitation and maintenance of this kind of effector response can provide guidance for vaccine design. In this review, we discuss the evidence for CD8+ T cells as correlates of protection, the means by which their antiviral capacity is evaluated, and transcription factors responsible for their function, or dysfunction, during HIV infection.
Keywords: T cells, cytotoxicity, cytokines, transcription factors, HIV
Introduction
A recent UNAIDS report estimates that there are currently 34 million people infected with human immunodeficiency virus (HIV), the causative agent of acquired immunodeficiency syndrome (AIDS)(1). With almost 2 million deaths each year from AIDS-related illnesses, HIV/AIDS remains one of the leading causes of death globally. While extensive prevention education initiatives and therapeutic intervention have contributed to reducing incidents of HIV infection and mortality over the last decade, the number of new infections remains high at more than 2 million new infections annually (1). These data indicate that there continues to be a pressing need for the development of an effective vaccine to control the HIV pandemic.
There are three potential strategies for a HIV vaccine design: a humoral approach, a cellular approach, or a combination of the two. While initial attempts to induce protection via humoral responses (AIDSVAX B/B and AIDSVAX B/E) or cellular responses (STEP trial) provided little to no protection (2–4), the Thai RV144 vaccine trial, which aimed to elicit both humoral and cellular immunity, met with partial success and provided the first evidence that it may be possible to protect against HIV acquisition (5). However, the RV144 vaccine strategy failed to elicit strong neutralizing antibody activity or CD8+ T-cell responses (6, 7). In addition, the protective effects were modest and of limited durability, and there is some question as to whether the results are generalizable to groups with greater risk of HIV acquisition than the cohort examined in the RV144 trial (8). It will take several years to determine if the RV144 ALVAC-HIV/AIDSVAX B/E approach is truly protective or can be improved upon, and it is important that alternate vaccine strategies be pursued to either complement the ALVAC-HIV/AIDSVAX B/E vaccine or replace it in the case of its failure.
A truly efficacious HIV vaccine will include the induction of antiviral cellular responses mediated by CD8+ T cells. As such, a detailed understanding of the properties of CD8+ T cells that correlate with virologic control is essential to focus vaccine development on strategies that will elicit appropriate cellular responses. The recent failure of the Merck STEP trial would appear to suggest that CD8+ T-cell responses do not prevent infection nor lower viral set-point following infection (9). However, there is substantial correlative evidence indicating CD8+ T-cell responses play a significant role in controlling HIV infection at some level, if not completely (10–16). Additionally, recent data from preclinical rhesus macaque studies suggests that CD8+ T cells induced by vaccination can indeed provide some degree of protection from SIV infection (17–20), supporting the idea that if strong responses in the right state of activation and anatomical location can be induced they could be effective. Together, these studies tell us we must extend our understanding of CD8+ T-cell responses beyond the examination of a single function, such as IFN-γ, to define the optimal measures on which to infer vaccine efficacy.
In this review, we discuss the various CD8+ T-cell functions elicited during infection, potential correlates of protection, and transcription factors involved in CD8+ T-cell differentiation and effector function. Understanding the responses that are associated with control of HIV disease progression as well as the complex network of transcription factors involved in CD8+ T-cell differentiation may identify useful targets for development of therapeutics as well as vaccines.
Cytotoxic CD8+ T-cell function
Antigen-specific CD8+ T cells are a heterogeneous population capable of performing multiple functions. Several studies have demonstrated this heterogeneity in the responses to HIV-1, cytomegalovirus (CMV), and Epstein-Barr virus (EBV) infections (21–25). These responses include production of cytokines and chemokines, cytolytic effector molecules, and antigen-specific lysis of major histocompatibility complex (MHC) class I matched target cells (Fig. 1). The majority of responding CD8+ T cells exert multiple functions following stimulation, but they can also respond with as little as one (depending on the number of parameters measured). Many of these functions are readily detectable by enzyme-linked immunospot (ELISpot) assay or flow cytometry and play specific, and potentially differential, roles in immunity against a variety of viruses. Several of the functions associated with CD8+ T cells that are frequently assessed in response to HIV infection are described in brief below.
Fig. 1. Multifunctional nature of the CD8+ T cell-mediated HIV antiviral response.
Schematic of CD8+ T cell (blue) interacting with HIV-infected or uninfected CD4+ T cells (red). CD8+ T cells directly inhibit replication by perforin and granzyme-mediated lysis of infected cells. They also promote a generalized antiviral state through secretion of cytokines (IFN-γ and TNF-α), prevent HIV transcription in infected cells by as yet unidentified molecules (CAF), and block HIV infection of target cells through β-chemokines (MIP-1α, MIP-1β, and RANTES).
Interferon-γ
Interferon-γ (IFN-γ) is the only member of the type II class of interferons, a family of cytokines originally discovered for their ability to interfere with influenza virus replication (26). IFN-γ is the single most commonly used function to assess CD8+ T cell responses to infection or vaccination. It promotes a general antiviral state by inducing the conversion of the constitutive proteasome to the immunoproteasome (27), upregulating expression of the TAP transporter proteins (28, 29), and increasing expression and stability of MHC class I molecules (30, 31). In some contexts, IFN-γ also increases the susceptibility of virally infected cells to apoptosis by increasing the expression of the TNF-α receptors and Fas/FasL (32, 33). However, IFN-γ may also enhance HIV replication (34). Thus in the context of HIV infection, IFN-γ is potentially both beneficial and detrimental to inhibiting viral replication.
Interleukin-2
Interleukin-2 (IL-2) is the primary growth factor for T cells (35). Although typically considered a CD4+ T-cell cytokine, CD8+ cells are also quite capable of producing IL-2 (36). It has no direct antiviral effector function, but it does promote expansion of CD4+ and CD8+ cells, thereby amplifying the effector response to pathogens (37). IL-2 may also be important for programming CD8+ T cells for better memory generation and effector function (38). IL-2 production by CD8+ T cells is correlated with proliferation of CD8+ cells independent of CD4+ T-cell IL-2 production (36), and both IL-2 production and proliferation are preserved in nonprogressive HIV infection (39, 40). However, IL-2-induced activation and proliferation of CD4+ T cells may also increase the availability of target cells for infection as well as increase viral replication by infected cells (41).
Tumor necrosis factor-α
Tumor necrosis factor-α (TNF-α) is a member of the TNF superfamily and was first identified by its ability to induce necrosis in solid tumors (42). It has subsequently been shown to be an important antiviral factor, due to its role as a mediator of apoptosis as well as inflammation and immunity (43–45). TNF-α is initially expressed as a biologically active homotrimer on the cell surface that the matrix metalloprotease TNF-α converting enzyme can subsequently cleave into its soluble form (46). Soluble TNF-α preferentially binds TNF-RI, which, upon being bound, initiates a signaling cascade that induces apoptosis of infected cells. Membrane-bound TNF-α binds TNF-RII and plays an important role in driving NF-κ B activation and inflammation (47, 48). TNF-α also promotes an antiviral state by enhancing expression of MHC class I and by inducing expression of IL-12 and IL-18, which are both important for upregulating production of IFN-γ by CD8+ T cells (31, 49, 50). However, similar to IFN-γ and IL-2, activation of cells induced by TNF-α can also result in increased production of virus (51–53).
Cytotoxic inhibitory mechanisms
Perhaps the most important function of CD8+ T cells is to recognize and kill infected cells. This function has been shown to be important for control of several infections, including EBV (54), CMV (22, 55), hepatitis B virus (HBV) (56), and hepatitis C virus (HCV) (57). CD8+ T cells predominantly mediate killing through the secretion of granzymes and perforin (58, 59). Granzymes are serine proteases that cleave caspases to induce apoptosis (60, 61), and perforin is a pore-forming protein that is required for delivery of granzymes into a target cell (62, 63). Both of these proteins are contained within lytic granules and are released early after CD8+ T-cell activation into the immunological synapse formed between the CD8+ T cell and a target cell. This process of degranulation is MHC class I restricted and antigen specific and likely plays an important role in control of viral infection in vivo (64).
CD8+ T cells can also mediate killing by the Fas-Fas ligand (FasL) pathway. FasL is upregulated by CD8+ T cells following activation by a target cell (65). Cross-linking of membrane bound FasL and the cell surface death receptor Fas expressed on targets cells induces assembly of an intracellular death-inducing signaling complex (DISC) (66). DISC formation causes activation of a caspase cascade that ultimately leads to apoptosis of the target cell. Individual CTL are thought to be capable of both FasL- and perforin-mediated killing (67); however, cytolysis of HIV-infected target cells appears to be largely perforin-mediated with no clear evidence of a contribution of FasL-mediated killing by HIV-specific CD8+ T cells (59). In addition, reports that a soluble form of FasL can not only block apoptosis but also induce proliferation and NF-κ B activation of HIV target cells raises the possibility that its role in infection is not always directly antagonistic (68, 69).
Noncytotoxic inhibitory mechanisms
Both CD4+ and CD8+ T cells secrete a variety of chemotactic cytokines (chemokines) upon activation (70). Chief among them are the β-chemokines macrophage inflammatory protein-1α (MIP-1α) and MIP-1β and regulated upon activation normal T-cell expressed and secreted (RANTES). MIP-1α and MIP-1β can be found in cytotoxic granules, while RANTES is stored in a separate secretory compartment called the RANTES secretory vesicle (RSV) (71, 72). Both types of granules are rapidly released following T-cell activation. New MIP-1α and MIP-1β synthesis occurs within a few hours of activation, while RANTES can take several days to be upregulated following its initial release. All three contribute to an inflammatory response primarily by recruiting leukocytes to the site of injury or infection.
β-chemokines were the first noncytotoxic factors secreted by CD8+ T cells to be identified that directly inhibit HIV replication (73). They inhibit replication in vitro by binding their cognate chemokine receptor, CCR5, which serves as a coreceptor for viral binding and entry into target cells. Binding of β-chemokines to CCR5 is thought to block access to and induce the internalization of the receptor (74). The exact role the β-chemokines play during HIV infection in vivo may still be a matter for debate. β-chemokines do not appear to prevent infection of monocytes and may actually enhance viral replication in these cells (75–77). RANTES (but not MIP-1α or MIP-1β) can increase attachment of HIV to cells in a manner independent of both CD4 and CCR5 and increase replication by activating signal transduction pathways (78). Serum β-chemokine concentrations do not correlate with HIV disease status, as patients with progressive infection tend to have higher levels than those with non-progressive infection (79). There is also the suggestion that physiologic levels of β-chemokines are not high enough to exert anti-HIV activity (80), although there is the possibility that concentrations are sufficient for inhibition in the microenvironment of the CD8+ T cell. Thus, while these molecules have been shown to have inhibitory effects in vitro, they may in fact fuel infection in vivo by not only recruiting uninfected target cells to sites of active viral replication but also by enhancing infection of those cells.
Another noncytotoxic function, CD8+ T-cell antiviral factor (CAF) was originally defined in the context of HIV infection, and the demonstration of its activity provided the first indication that CD8+ T cells possess the ability to inhibit HIV replication (81). CAF is a diffusible lymphokine that lacks identity with IFN-α, IFN-β, TNF-α, IL-4, IL-6, or the β-chemokines MIP-1α, MIP-1β and RANTES (82–85). Aside from this, there is little known and much debate about the exact nature of CAF (83, 86, 87). It may be the activity of one or more cytokines or chemokines acting together, or it could be an as yet unidentified molecule (88). In the case of HIV, CAF appears to function by suppressing HIV long terminal repeat (LTR)-mediated gene expression in CD4+ T cells (89). It does not block HIV entry (89), proviral integration (90), or reverse transcription (87), nor is it MHC class I restricted (86). Due to CAF activity being neither HIV-antigen specific nor produced only by CD8+ T cells, it has been hypothesized that it may in fact be part of an innate rather than an adaptive immune response (88, 91). Despite this, the suppressive capacity of CAF appears to be real, and further investigation is warranted to determine its identity.
Work from two separate groups provides evidence of a significant role for noncytolytic inhibitory mechanisms in viral control during chronic infection. Wong et al. (92) and Klatt et al. (93) both used a combination of experimental depletion of CD8+ T cells and ART treatment in SIV-infected rhesus macaques to examine the in vivo effect of CD8+ T cells on lifespan of productively infected cells. They each found the estimated decay rate of infected cells is similar in the presence or absence of CD8+ T cells, suggesting clearance of infected cells by CD8+ T-cell cytotoxic mechanisms does not account for the entirety of CD8+ T-cell-mediated control during chronic infection. However, whether control is the result of CAF, β-chemokines, or some other noncytolytic function has not been determined. These studies also do not rule out the possibility of a greater role for cytotoxic effects in the context of acute or controlled infection.
CD8+ T cells and correlates of protection
Evidence that cytotoxic CD8+ T lymphocytes (CTL) are important for control of HIV replication comes from both HIV infection in humans and simian immunodeficiency virus (SIV) infection in nonhuman primates (NHP). First, the resolution of peak viremia during acute HIV infection is temporally associated with the expansion of HIV-specific CD8+ T cells (10, 11). Second, immunologic pressure exerted by HIV- and SIV-specific CD8+ T cells is linked to the emergence of viral escape mutations during acute and chronic infection (12–14, 94, 95). Third, there is a strong correlation between specific MHC class I alleles and non-progressive infection in both humans and rhesus macaques (15, 16, 96–98). Finally, experimental depletion of CD8+ T cells in SIV-infected rhesus macaques results in a concomitant loss of control of viral replication (99, 100).
Despite a clear role for CD8+ T cells in the initial control of HIV replication, a correlate of protection has remained elusive. Much of the research in the field has focused on HIV seropositive individuals who maintain normal CD4+ T-cell counts and are clinically healthy for 10 or more years [long-term nonprogressors (LTNPs)] and those who control viral replication to below the limit of detection (elite controllers or ECs). Although the initial immune responses in these individuals fail to prevent infection, by determining the nature of the responses that subsequently control HIV replication we may better inform vaccine designs that are therapeutic if not prophylactic. Studies seeking to define the mechanism(s) of control have largely relied on the comparison of HIV-specific CD8+ T-cell responses from ECs to those from chronic progressors (CPs). Characterization of these responses has evolved over the past two decades and has provided some of the first clues to cellular mechanism(s) of control.
Early work in the field employed cytotoxicity assays, such as the chromium release assay (CRA), to measure HIV-specific CTL activity. The CRA was used to show that HIV-specific CD8+ T cells have direct cytotoxic effects against HIV-infected CD4+ T cells (101) and was important in establishing the link between emergence of HIV-specific CTL and resolution of peak viremia during primary HIV infection (10). However, as discussed later, questions were raised about the ability of the assay to link CTL activity and HIV viral load during chronic infection (102). The CRA is also laborious, relatively insensitive, highly variable, and, most importantly, provides little information about the cytolytic CD8+ T cells themselves other than that they can kill.
Direct detection and quantification of antigen-specific CD8+ T cells by MHC class I tetramer technology or IFN-γ production offered more rapid, more sensitive, and less variable assays than measurement of cytotoxicity by the CRA. While an inverse relationship between simple frequency of HIV-specific CD8+ T cells and plasma viral load was initially established on the basis of tetramer staining (102), this finding was not supported by subsequent studies that found no relationship between the frequency of IFN-γ-producing HIV-specific CD8+ T cells and HIV viral load (103–105). It was proposed that these disparate findings were the result of a significant portion of the circulating tetramer-staining CD8+ T-cell population being functionally impaired (106, 107). While early studies in this area have shown that the majority of tetramer-positive HIV-specific CD8+ T cells can produce IFN-γ (23, 108), high expression of inhibitory markers including PD-1, CD160, 2B4, and Lag-3 on HIV-specific CD8+ T cells may indicate some degree of functional insufficiency (109–113).
IFN-γ was presumed to be an antiviral marker largely because CD8+ T-cell clones that produced it early after stimulation became CTLs after further culture (114, 115). This observation, combined with its ease of measurement by ELISpot or flow cytometry, made IFN-γ a popular choice for detecting HIV-specific cellular responses in both HIV-infected individuals and participants in HIV vaccine trials. However, several studies have demonstrated the inadequacy of IFN-γ as a surrogate marker for HIV control. One study found a positive correlation between frequency of IFN-γ-producing HIV-specific CD8+ T cells and HIV viral load (105), while two others showed that although HIV-specific CD8+ T cells from ECs and CPs can both produce IFN-γ, IFN-γ-producing CD8+ T cells from CPs are impaired in both cytotoxicity and proliferative capacity compared to cells from ECs (40, 116). The failure of the STEP trial further highlighted the misinterpretation of IFN-γ as a surrogate of protection: 77% of vaccinees had an IFN-γ response to one or more HIV antigens by ELISpot, yet there was no protection from infection or enhanced viral control following infection (9). While increased rates of escape mutations within vaccine-targeted CD8+ T-cell epitopes indicate that vaccine-induced immune pressure was exerted on the virus, the nature of the selective forces has not been defined (117). More recently, studies have shown more directly that the magnitude of IFN-γ responses does not correlate with CD8+ T-cell HIV inhibitory activity (116, 118). Thus, while IFN-γ may be a good indicator of the presence of a response, it cannot be used alone to infer the anti-HIV capacity of T cells.
Polyfunctional T cells
As discussed earlier, antiviral CD8+ T-cell responses have the capacity to produce several different functions, and measuring a single function such as IFN-γ likely does not describe the true extent of an antigen-specific immune response. This idea was confirmed by studies based on the murine lymphocytic choriomeningitis virus (LCMV) model of infection. The Armstrong strain of LCMV is cleared following acute infection, while the clone 13 strain results in chronic infection. Following resolution of acute infection by the Armstrong strain, a subset of LCMV-specific IFN-γ-producing CD8+ T cells emerges that also produces IL-2 (119). This same subset does not appear following chronic infection with LCMV clone 13, as LCMV-specific cells continue to produce only IFN-γ. Wherry et al. (120) extended these findings to show that CD8+ T cells are not only multifunctional but also that functional capacity of LCMV-specific CD8+ T cells is gradually lost in the context of chronic clone 13 infection. Cells producing IL-2 were the first functional subset lost, followed by those producing TNFα, while IFN-γ was the most resistant to this ‘functional exhaustion’. These studies demonstrate that individual CD8+ T cells are capable of responding with multiple functions simultaneously and that measuring IFN-γ alone fails to exclude T cells that are potentially impaired in their functional capacity. They also indicate that including more than one functional marker during the assessment of cellular responses to infection or vaccination will identify cells with greater antiviral potential.
Appay et al. (23) and Sandberg et al. (121) were the first to demonstrate the functional complexity of human CD8+ T-cell responses. They examined cellular responses to either HIV and CMV (23) or CMV alone (121) and showed that different cells specific for the same antigen were capable of producing TNF-α, IFN-γ, and MIP-1β or IL-2, TNF-α and IFN-γ, respectively. Technical limitations prevented either of these studies from examining the capacity of individual antigen-specific cells to co-produce cytokines or chemokines; however, advances in flow cytometry technology allowing the concurrent measure of up to 18 functional and phenotypic markers soon provided a new tool for assessment of responses (122). The first true demonstration of multifunctional T cells in humans came when De Rosa et al. (123) examined responses of antigen-specific T cells elicited by HBV-and HIV-vaccination or natural infection, measuring five different functions (IL-2, TNF-α, IFN-γ, MIP-1β, and IL-4) simultaneously. Both CD4+ and CD8+ T cells displayed a surprising breadth and complexity of responses that could not have been captured by the measurement of any single function alone. Antigen-specific CD8+ T cells were capable of producing IL-2, TNF-α, IFN-γ, and MIP-1β alone or in combination. Importantly, there were several functional subsets that did not include IFN-γ production and thus would have been missed had IFN-γ been measured alone. This study established that measurement of multiple functions provides a more sensitive and complete evaluation of T-cell responses elicited by vaccination or natural infection. It also presented the possibility that distinct functional expression patterns might provide correlates of protection or disease progression.
Historically, neither the quantity nor the breadth of the HIV-specific CD8+ T-cell response has correlated with protection. However this is based almost entirely on analysis of IFN-γ responses alone, which is likely not representative of the true response to infection. Given the capacity of CD8+ T cells to produce multiple functions, it is possible that the key to protection is a matter of quality of the response rather than simply quantity. Zimmerli et al. (36) were the first to show that higher quality HIV-specific CD8+ T-cell responses consisting of simultaneous production of IL-2 and IFN-γ, compared to IFN-γ alone, differentiate nonprogressive and progressive HIV infection, respectively. The ability of nonprogressor CD8+ T cells to produce IL-2 was consistent with the maintenance of greater proliferative capacity than for cells from progressors (36, 40). These data suggested that quality of the response was indeed important. To investigate this association further, our laboratory measured five T-cell functions (IL-2, IFN-γ, TNF-α, MIP-1β, and CD107a) simultaneously to characterize HIV-specific CD4+ and CD8+ T-cell responses in a cohort of progressors and nonprogressors (39). This study demonstrated that while the absolute frequency of HIV-specific CD8+ T cells does not correlate with viral control, the frequency of specific functional subsets does correlate with control. Comparing the functional profiles of the HIV-specific CD8+ T-cell responses between progressors and nonprogressors, the two groups were differentiated by a higher degree of functionality in the nonprogressors than in the progressors. A population of CD8+ T cells capable of producing all five functions was observed almost exclusively in the nonprogressors, and a subset of CD8+ T cells producing IFN-γ, TNF-α, MIP-1β, and CD107a was also more prevalent in this group than in progressors. These responses were consistent across multiple HIV proteins (Gag, Pol, Env and Tat/Rev/Vif/Vpr/Vpu). Furthermore, within the progressor group, the magnitude and proportion of the HIV-specific CD8+ T-cell responses positive for these same five- and four-function subsets inversely correlated with viral load. This demonstrates that by measuring multiple functions simultaneously it is possible to discern a difference between progressive and nonprogressive HIV infection based on the functional capacity of HIV-specific CD8+ T-cell responses.
In support of an association between polyfunctional cells and protection during infection, Darrah et al. (124, 125) demonstrated that vaccine-elicited polyfunctional CD4+ T cells are far better at providing protection to mice challenged with Leishmania major than dualfunctional or monofunctional cells. However, a similar phenomenon for CD8+ T cells has not been established in this or other models of infection, and the role of polyfunctional CD8+ T cells in the context of HIV infection is surrounded by questions. First, how can polyfunctionality be a significant correlate of protection, when only a small proportion of nonprogressor HIV-specific CD8+ T cells actually produce five functions and some nonprogressors do not have this or any other measurable functional subset (39, 126)? Second, why do polyfunctional HIV-specific CD8+ T cells induced in progressors during antiviral therapy generally fail to impart virologic control upon cessation of therapy (127–129)? Finally, how do IL-2, TNF-α, IFN-γ, and MIP-1β become protective in combination when they are not individually associated with protection and they all have potentially pleiotropic effects in the context of HIV infection (as discussed earlier)? Is it a synergistic effect of multiple functions acting in concert, or the ability of polyfunctional antigen-specific CD8+ T cells to produce more cytokine on a per cell basis (130, 131)? Is it simply that these cells are functional in a way not yet measured? All of these issues come down to a single question: is polyfunctionality a cause or consequence of control? Polyfunctionality, as defined by the above measures, is not indicative of the protective capacity of a cell, and their use as a surrogate for a protective response in vaccine studies is not yet founded on direct evidence that they themselves mediate control.
Cytotoxicity and perforin
Cytotoxicity is one function of CD8+ T cells that is unequivocally antiviral, as direct killing of virally infected cells will certainly impact viral replication. Cytotoxic capacity can be assessed directly or indirectly by several different methods, each with its own advantages and disadvantages. As discussed earlier, direct cytotoxic capacity of CTLs can be assessed in vitro using the chromium release assay or by a similar fluorescence-based assay, and these assays were important for establishing the role of CTL in controlling HIV infection (10, 11, 101). Although several studies have since used the CRA to show HIV-specific CD8+ T cells from ECs maintain greater cytotoxicity than those from CPs (23, 132, 133), the connection between the results of these assays and the in vivo state has been questioned (102). Critics of this interpretation cite the prolonged culture and expansion of cells in vitro required to generate sufficient numbers of CTLs for quantitative CRA (134), which likely introduces bias in CTL analysis as it only measures the subset of memory and effector cells capable of proliferating and remaining functional under artificial conditions. Additionally, these assays only measure the effects of CTL on the target cells, without providing any phenotypic or functional characteristics about the CTL itself. This represents a critical loss of information when trying to determine the exact nature of these cells.
CD8+ T-cell cytotoxic potential can also be assessed directly by flow cytometry. Unlike the cytotoxicity assays, this method does not measure killing but rather determines killing potential based on the perforin and granzyme content of the CD8+ T cell, with the concept that CD8+ T cells that do not express these markers would be incapable of killing targets. The advantage to this method is that flow cytometry permits measurement of additional parameters, thus providing a more complete picture of the cells with killing potential. Perforin content of antigen-specific CD8+ T cells can be measured by staining resting cells with MHC class I tetramers or by peptide-stimulation. When Appay et al. (23) compared perforin content of CP antigen-specific CD8+ T cells identified directly ex vivo by tetramer staining, they found that HIV-specific CD8+ T cells were deficient for perforin compared to CMV-specific CD8+ T cells. However, MHC class I tetramer staining may provide sufficient stimulation to induce perforin release, resulting in an underestimation of perforin content. Also, by measuring perforin content of resting cells, only immediate killing potential is measured, and the sustainability of the cytotoxic response cannot be determined. To assess serial killing potential, CTLs had to be stimulated and perforin content measured after the cells were allowed to progress through multiple rounds of proliferation. Several studies measuring cytotoxic potential in this way suggested that HIV-specific CD8+ T cells from ECs maintain the ability to upregulate perforin, a property that was deficient in CPs (23, 40, 133). While these results were in line with the results from the cytotoxicity assays, they also shared the same potential for memory subset bias introduced by the need for prolonged culture.
As a result of the difficulty in measuring cytotoxic factors in activated cells directly ex vivo, we developed an assay that utilized CD107a expression on CD8+ T cells as a surrogate marker of killing capacity (135). CD107a lines the membranes of cytotoxic granules and is exposed on the cell surface following activation and degranulation of CD8+ T cells. This technique was used recently to suggest cytotoxicity is one of the primary mechanisms responsible for both the enhanced HIV inhibitory potential of CD8+ T cells from ECs as well as for the emergence of HIV escape mutants (136–138). It is important to note, however, that although expression of CD107a correlates well with the exocytosis of perforin and cytotoxicity (135, 139), it does not directly equate to actual killing, as not all granules contain perforin or granzymes (140).
Through further testing, we soon discovered that the most commonly used perforin antibody (clone δG9) was sensitive to the degranulation assay, which required the use of monensin to neutralize intracellular pH (139). By using a different monensin-insensitive perforin antibody (clone D48), we found that CD8+ T cells are capable of rapidly (within 4–6 h) upregulating perforin following antigen-specific stimulation without the requirement for proliferation (139, 141). We also found that newly synthesized perforin can traffic directly to the immunological synapse in a process that largely bypasses cytotoxic granules (141). This new perforin antibody thus allowed a direct assessment of antigen-specific CD8+ T-cell cytotoxic potential directly ex vivo. Used in conjunction with standard intracellular cytokine staining (ICS), we could now assess cytokine production and cytotoxic potential simultaneously.
Perforin upregulation and polyfunctionality
In an attempt to better understand the HIV-specific CD8+ T-cell mechanisms of viral control, we re-evaluated HIV-specific CD8+ T-cell polyfunctional responses, including the ability to rapidly upregulate perforin. Comparing the functional profiles of HIV-specific CD8+ T-cell responses from a cohort of elite controllers and chronic progressors, we confirmed that elite controllers have a higher frequency of polyfunctional cells compared to chronic progressors based on the expression of IL-2, IFN-γ, TNF-α, MIP-1β, and CD107a (142). HIV-specific CD8+ T cells from elite controllers also have a greater ability to rapidly upregulate perforin directly ex vivo compared to progressors. This identifies a function of the elite controller HIV-specific CD8+ T-cell response that confers an enhanced ability to directly eliminate HIV-infected cells. Interestingly, however, there was no link between perforin upregulation and polyfunctionality. Rather, perforin expression was associated with cells of limited functional capacity, most frequently seen in combination with production of MIP-1α or degranulation (CD107a). Thus, while highly polyfunctional cells are a correlate of control, so too are oligofunctional cells, depending on the functions produced. The question then became which of these populations, if either, is involved in control of viremia. Freel et al. (138) recently demonstrated that HIV-specific CD8+ T-cell capacity to inhibit HIV replication in vitro is correlated with the expression of MIP-1β and CD107a and not linked to upregulation of IL-2, TNF-α, or IFN-γ. While noncytolytic mechanisms likely play some role in inhibition (136, 143), given our findings it is not unreasonable to speculate that many of the cells expressing MIP-1β and CD107a are also upregulating perforin and inhibiting mainly through cytotoxicity, although this hypothesis has not been confirmed. Still, these studies only establish that HIV-specific CD8+ T cells found during controlled HIV infection have greater functional capacity and inhibitory potential, they do not tell us if these responses are actively involved in virus control or they are a result of control by some other mechanism.
CD8+ T-cell responses in acute HIV infection
HIV-specific CD8+ T cells are elicited early following HIV infection and are associated with control of viremia as well as selection of escape mutants (13, 144, 145). As the long-term control of HIV is likely determined by CTL responses during the resolution of acute viremia, we decided to characterize the functionality of HIV-specific CD8+ T cells during this phase of infection. It stands to reason that if polyfunctionality or rapid perforin upregulation are responsible for control, they should be present during this critical time period. Similar to other reports (137, 146, 147), when we examined expression of IL-2, IFN-γ, TNF-α, MIP-1α, and CD107a, we found that HIV-specific CD8+ T cells are rarely polyfunctional during acute infection with the highest frequencies of responding cells represented by the expression of MIP-1α and CD107a singly or in combination (Makedonas G, Betts M, et al., manuscript in preparation). In contrast to previous reports indicating perforin expression during acute infection is low (148, 149), we found that rapid perforin upregulation dominates the earliest HIV-specific CD8+ T-cell responses we could detect in nearly every acute subject (Makedonas G, Demers K, Betts M, et al., manuscript in preparation). This finding suggests that the appearance of highly polyfunctional HIV-specific CD8+ T cells observed in ECs is likely a result of viremic control, while a robust perforin-mediated response may be responsible for clearance of acute viremia. This idea is supported by a recent study comparing the HIV-specific CD8+ T-cell responses of ECs with individuals who initiate antiretroviral therapy immediately following infection (150). In this study, early, long-term therapy resulting in control of viremia led to the establishment of polyfunctional HIV-specific CD8+ T-cell responses that were identical to controller responses. These data suggest that polyfunctional HIV-specific CD8+ T cells emerge as a result of effective control of HIV replication while CD8+ T-cell cytotoxicity mediated by perforin expression is likely responsible for viral control.
CD8+ T-cell responses in gut mucosa and lymph nodes
The vast majority of HIV replication takes place in mucosal and lymphoid tissues, and it is not clear that HIV-specific CD8+ T-cell responses found in the blood are representative of responses found at these sites. As such, we need to broaden our understanding of CD8+ T-cell function within relevant tissues to pinpoint correlates of protection. Critchfield et al. (151) and Ferre et al. (131) both found associations between polyfunctional responses and disease status investigating production of IL-2, IFN-γ, TNF-α, MIP-1β, and CD107a by HIV-specific CD8+ T cells from human gut mucosa. Although there was a paucity of cells producing all five functions, an IFN-γ/TNF-α/MIP-1β/CD107a population clearly segregated controllers from noncontrollers, a finding largely in agreement with our observations from peripheral blood cells. However, Shacklett et al. (152) found little to no resting perforin expression by CD8+ T cells from rectal mucosa of both chronically infected HIV-positive and healthy individuals. Whether this finding is the same when looking at upregulated perforin or extends to the gut mucosa of ECs as well will need to be determined by more comprehensive studies comparing ECs and CPs.
Data from the lymph nodes are less consistent. While Altfeld et al. (153) found a greater magnitude of HIV-specific CD8+ T cells in the lymph nodes than in peripheral blood, Connick et al. (154) demonstrated that CD8+ T cells have limited access to lymphoid follicles, the primary site of HIV replication within these tissues. Further, an early study by Sunila et al. showed activated perforin-expressing CD8+ T cells localized within follicles, whereas subsequent studies have found no perforin expression by lymph node-resident CD8+ T cells (133, 148, 155), particularly by those that do gain access to the follicle (155). The discrepancies between peripheral blood HIV-specific CD8+ T-cell responses and those from the gut or lymph nodes may be related to phase of infection during which samples were acquired, the progression status of the subjects studied, or simply a matter of differences in reagents used for analysis. It will be important to determine the true contribution of antiviral CD8+ T-cell responses at these sites given their role in HIV replication and dissemination. This is of particular importance given recent papers identifying lymph node CD4+ T-follicular helper cells as a new major reservoir of HIV infection (156–158).
Transcriptional regulation of CD8+ T-cell function and differentiation status
Accumulating evidence from our laboratory and others (40, 142, 159, 160) implicates the cytotoxic capacity of CD8+ T cells as a primary factor in the cellular control of HIV infection. Cytolytic potential is lost rapidly in most HIV-infected individuals, such that during chronic progressive infection only ~15% of HIV-specific CD8+ T cells express perforin, compared with ~40% in elite controllers (142). Understanding the underlying mechanisms involved in CD8+ T-cell differentiation and function may provide insight into the factors responsible for the dysfunction observed during chronic infection. To this end, several transcription factors have been identified that regulate the transition of CD8+ T cells into effector cells, including T-box expressed in T cells (T-bet), Eomesodermin (eomes), Runx3, and Blimp-1 (Table 1). While the majority of our current knowledge about the transcriptional control of CD8+ T-cell function comes from mouse models, we have recently begun assessing the role these transcription factors play in human T-cell effector function. Below, we discuss what we know from mouse models as well as our recent findings regarding CD8+ T-cell dysfunction in the context of HIV infection.
Table 1.
Transcriptional regulators of CD8+ T cell differentiation and function
| Transcription factor | Effect | References |
|---|---|---|
| T-bet | Cytotoxicity generation, granzyme and perforin production, terminal differentiation, represses PD-1 expression | 25, 165, 166, 167, 172, 174, 189 |
| Eomes | Cytotoxicity generation, granzyme and perforin production, expression of IL-15 receptor (CD122), upregulation of exhaustion markers | 169, 171, 172, 174, 181, 187 |
| Runx3 | CTL proliferation, Eomes production | 175, 176 |
| Blimp-1 | Cytotoxicity generation, granzyme and perforin production, terminal differentiation, represses Bcl-6 and Id3, upregulation of exhaustion markers | 177, 178, 179, 187 |
| Bcl-6 | Inhibits Blimp-1 and T-bet directly as well as their target genes | 180, 183, 184, 185 |
| Id2 | CTL expansion, terminal differentiation | 191, 192 |
| Id3 | Promotes survival during memory transition | 192, 193 |
| Notch1 | Eomes and perforin production | 194 |
| Notch2 | Granzyme B production | 195 |
| STAT3 | Sustains Bcl-6 and Eomes expression | 182 |
| STAT4 | T-bet expression and terminal differentiation | 196, 197 |
| STAT5 | Perforin production | 181, 198, 199 |
T-bet and Eomesodermin
T-bet and Eomes are both T-box binding transcription factors that play important roles in promoting CD8+ T-cell effector and memory differentiation and function. The T-box is a 180–190 amino acid sequence highly conserved across T-box family members that acts as a DNA-binding domain. T-bet and Eomes share 74% sequence identity in their T-box domains but lack sequence similarity outside this region. Like other T-box-binding transcription factors, they mediate developmental transitions through epigenetic modifications (161).
T-bet was originally identified as a determinant of T-helper 1 (Th1) cell lineage commitment (162), but subsequent work demonstrated its importance also as a regulator of CD8+ T-cell effector differentiation and function (163–165). T-bet positively regulates several genes associated with effector function, including perforin, granzyme B, β-chemokines, and IFN-γ (166), and negatively regulates genes that oppose effector function such as IL-2 and PD-1 (162, 166, 167). Although T-bet activity was originally described as being required for CD4+ and CD8+ T-cell IFN-γ expression and cytotoxicity, these functions were not completely ablated in T-bet knockout mice, suggesting the existence of a T-bet-independent regulatory mechanism (165, 168). This observation led to the discovery that CD8+ T cells also express Eomes, which shares both redundant and reciprocal functions with T-bet (169). The genes under the control of Eomes are not as well defined as for T-bet, but knockdown of Eomes in activated CD8+ T cells causes decreased expression of IFN-γ, perforin, and granzyme B, indicating there is at least some overlap with T-bet in promoting effector function. A combined T-bet and Eomes deficiency results in the loss of CTL identity and anomalous production of IL-17 by CD8+ T cells (170), suggesting that T-bet and Eomes coordinately regulate CTL differentiation.
T-bet and Eomes appear to play opposing roles in the generation of memory CD8+ T cells. Both transcription factors cause the upregulation of IL-2Rβ (CD122), which is required for long-term memory CD8+ T-cell survival and homeostatic proliferation in response to IL-15 signals (171). However, whereas high expression of Eomes correlates with central memory T (TCM) cells, high expression of T-bet represses IL-7Rα expression to drive formation of effector T and effector memory T (TEM) cell subsets at the expense of TCM cell generation (172, 173). T-bet is highest in early effector CD8+ T cells but progressively declines as memory cells form, while Eomes is initially upregulated in early effector CD8+ T cells but increases during the effector to memory transition (174). These divergent expression patterns may be partly explained by the way these transcription factors are induced. T-bet is rapidly induced in activated CD8+ T cells downstream of TCR signaling and augmented by inflammatory signals such as IL-12 (163, 164). Expression of T-bet initiates a positive feedback loop by upregulating IL-12Rβ, thereby increasing sensitivity to additional IL-12. Eomes induction is less well defined. However, studies indicate it is induced subsequent to T-bet, amplified by IL-2, and repressed by IL-12 (164). This suggests the differential expression patterns of T-bet and Eomes in CD8+ T cells, and in turn the differentiation state, depends on the degree of inflammation in the immediate environment of the cell.
Runx3
The runt domain-containing protein Runx3 is important for establishing the CD8+ T-cell lineage in the thymus, but its role in mature CD8+ T cells and mechanism of regulation are less clear. Previously, Runx3 knockout models showed reduced CD8+ T-cell cytolytic activity. This defect was later attributed to the role of Runx3 in driving CD8+ T-cell proliferation rather than a defect in cytolytic activity (175). More recently Cruz-Guilloty et al. (176) described a direct role for Runx3 in programming effector function, in cooperation with T-bet and Eomes, using in vitro differentiated CTLs. Similar to the findings described above, in this system, T-bet was induced early during differentiation, whereas Eomes was expressed at later stages. Interestingly, they showed only Eomes, not T-bet, could upregulate perforin, suggesting Eomes may be more important for CTL effector function. Importantly, Runx3-deficient CD8+ T cells failed to upregulate Eomes, Ifng, Gzmb (granzyme B), and Prf1 (perforin). However, while Runx3 was found to bind to the promoters for Ifng, Gzmb, and Prf1 in CTLs, it is remains unclear if Runx3 upregulates these effector molecules directly or via its induction of Eomes, which binds to the same promoters.
Blimp-1 and Bcl-6
The B-cell transcriptional repressor B lymphocyte-induced maturation protein-1 (Blimp-1) was first described for its ability to promote B-cell terminal differentiation into plasma cells. Three recent studies demonstrate that Blimp-1 plays a similar role in driving the terminal differentiation of effector CD8+ T cells over memory cells (177–179). These studies show that in antigen-specific CD8+ T cells, Blimp-1 expression increases during the acute response, has its highest expression in terminally differentiated CD8+ T cells, and decreases in memory cells following the resolution of infection, with the lowest expression levels observed in TCM cells. Blimp-1 is critical for normal perforin and granzyme B expression and promotes trafficking to sites of infection by upregulating the chemokine receptor CCR5 (178, 179). Like T-bet, Blimp-1 is induced by IL-12, but it is also induced by IL-2 and IL-21. Bcl-6 is crucial for the formation of TCM cells and is induced by IL-10 and IL-21 (180, 181). It is an antagonist of Blimp-1 activity, and its expression correlates inversely with Blimp-1 in effector and memory CD8+ T cells (182). Bcl-6 has also been shown to bind the T-bet promoter and T-bet itself in CD4+ T cells (183–185). While a similar interaction has not yet been demonstrated in CD8+ T cells, if confirmed, it would suggest Bcl-6 drives memory differentiation by repressing both Blimp-1 and T-bet directly, as well as the targets of these transcription factors.
Transcription factors in murine models of chronic infection
As discussed earlier, normal differentiation and function of virus-specific CD8+ T cells altered during chronic infection and exposure to persistent antigen. Effector cells fail to form functional memory populations and instead gradually become more exhausted. A few transcription factors have been shown to play a role in this process, including Blimp-1, T-bet, and Eomes (167, 177, 186, 187). Blimp-1 drives effector differentiation during acute infection; however, in the context of chronic infection, Blimp-1 also promotes the expression of the inhibitory receptors PD-1, CD160, 2B4, and Lag3, thereby inducing an exhausted state (177). T-bet sustains virus-specific CD8+ T-cell responses during chronic infection and suppresses the expression of PD-1. As T-bet expression decreases over time during chronic infection, PD-1 levels and CD8+ T-cell dysfunction both increase (167). Eomes also sustains virus-specific CD8+ T-cell responses during chronic infection, but its expression is also associated with a more terminally differentiated state, with high PD-1 expression levels, and Blimp-1 expression, which combined, indicate Eomes also plays a role in promoting exhaustion (187). Thus, the same transcription factors responsible for driving effector CD8+ T-cell differentiation and memory formation during acute infection are also responsible for limiting responses and preventing immunopathology in the context of chronic infection. Any attempt to manipulate these factors for purposes of immunotherapy will have to take these dual roles into account so that potential functional gains are not negated by simultaneously increasing terminal differentiation and exhaustion.
Transcriptional regulation of human CD8+ T cells
Although the molecules described above have been studied extensively in mouse models, much less is known about them in the context of human immunology. Investigation of transcription factors in human lymphocytes at the single cell level is largely limited to T-bet and Eomes as poor reagent availability precludes the assessment of Runx3 or Blimp-1. In the first report to fully characterize the expression of T-bet and Eomes in healthy human T cells, we found that Eomes is typically expressed with T-bet in CD8+ T cells and, in most cases, is bimodally distributed (188). T-bet, on the other hand, has a distinct trimodal expression pattern with a clear intermediate (or T-betlow) population in the majority of individuals. We also found that differential T-bet expression levels are associated with specific functional characteristics: T-betlow and negative cells are more likely to express IL-2 compared to T-bethigh cells, which tend to express perforin and granzyme B (25). These data are in agreement with data from mouse models in which T-bet promotes perforin and granzyme B expression, while repressing IL-2 production. T-bet and Eomes expression patterns also closely associate with memory phenotype: naive cells express little to no T-bet; TCM cells express a little of both; there are more TEM cells expressing T-bet overall, but with low MFI; and effector T cells express high levels of T-bet, with a large population of cells having high T-bet MFI. Eomes association with memory phenotype in healthy humans diverges somewhat from the mouse model, although there may be instances in human infections where these may be more similar (187). We found that significantly more cells in the effector T and TEM subsets express Eomes than in the TCM subset and, in the case of TEM cells, Eomes is also expressed at a higher MFI. Polychromatic imaging cytometry (Amnis ImagestreamX) analysis of the T-bethigh and T-betlow cells revealed an important difference in the localization of T-bet in human T cells; nuclear localization was generally observed in T-bethigh cells cytoplasmic localization was more common in T-betlow cells (188). Looking at T-bet expression levels and localization in memory populations, TCM cells generally express low levels of cytoplasmic T-bet, whereas effector T cells tend to express higher levels of nuclear T-bet. Thus while a cell may contain T-bet, if it is not expressed at high enough levels it remains sequestered in the cytoplasm of the cell and is transcriptionally inactive.
T-bet expression by HIV-specific CD8+ T cells
Having previously shown that HIV-specific CD8+ T cells from chronic progressors have a reduced ability to upregulate perforin compared to ECs, we examined the potential role of T-bet in driving effector function in each of these two groups (189). Similar to the expression pattern observed in normal healthy donors, perforin and granzyme B were closely associated with T-bethigh CD8+ T cells in the context of HIV infection. More importantly, we demonstrated that T-bet is significantly lower in HIV-specific CD8+ T cells from CPs compared to ECs. A recent report by Ribeiro-Dos-Santos et al. (190) confirmed our results through T-bet (and eomes) mRNA expression, finding reduced T-bet mRNA production during chronic HIV infection.
Given our recent findings regarding T-bet localization in human CD8+ T cells, the differential T-bet expression levels observed in the context of HIV control suggests that T-bet in elite controller HIV-specific CD8+ T cells may be predominately nuclear and therefore able to drive the effector gene cassette, while T-bet in chronic progressor HIV-specific CD8+ T cells is cytoplasmic and cannot upregulate effector function. If the association between localization and T-bet expression levels is confirmed in HIV-specific CD8+ T cells, these observations will provide a tangible mechanism to explain defects in the antiviral activity of HIV-specific CD8+ T cells in chronic progressors. Additionally, it will be important to determine when during the infection course this defective expression pattern is established. Two potential models are (i) HIV-specific CD8+ T cells from both ECs and CPs express T-bet at high levels during acute infection but failure to control viral replication results in loss of the T-bethigh population in CP, or (ii) EC HIV-specific CD8+ T cells are better able to express high levels of T-bet from the outset thereby providing a more robust antiviral response and better control.
It will also be important to look at the role of other transcription factors in the context of acute and chronic HIV infection, as T-bet does not act alone to regulate CD8+ T-cell differentiation and function (Table 1). For instance, Paley et al. (187) recently examined the expression of both T-bet and Eomes in HCV-specific CD8+ T cells from individuals who were either chronically infected with HCV or who had spontaneously cleared the infection. HCV-specific CD8+ T cells during chronic infection had higher levels of Eomes and relatively low levels of T-bet compared to HCV-specific CD8+ T cells from cleared HCV infection. This result mirrored their findings in a murine model of chronic LCMV infection. In addition, their murine model showed high Eomes expression was associated with high levels of Blimp-1 and the inhibitory receptors PD-1, Lag3, and Tim3, corresponding to a more exhausted phenotype. These data suggest that not only are low levels of T-bet unable to drive effector function in these cells, but Eomes is simultaneously promoting terminal differentiation and exhaustion which likely contributes to lack of control. Understanding the connection between CD8+ T-cell differentiation and function, and the complex network of transcription factors that regulates them, may provide a target for prophylactic or therapeutic strategies that lead to enhanced antiviral capacity in the face of chronic infection.
Concluding remarks
The identification of immune correlate(s) of protection will help form the basis on which to engineer and assess a prophylactic HIV vaccine. Although multiple studies have established the importance of measuring several T-cell functions simultaneously, the recent results of the Merck STEP trial emphasized the need to identify a true correlate of protection rather than relying on surrogate markers such as coproduction of IL-2, IFN-γ, and TNF-α. The ability of HIV-specific CD8+ T cells to clear infected cells through cytotoxic mechanisms represents the strongest indication of protective potential to date and should serve as a benchmark for cellular vaccine strategies. However, evaluation of additional functions remains important as noncytolytic mechanisms may also play a role in protection, and a multi-facetted assessment of responses offers the greatest odds of predicting vaccine efficacy. Additionally, understanding how novel factors, such as transcription factors, are involved in the regulation of CD8+ T-cell differentiation and effector function during acute and chronic infection may provide new targets for prophylactic or immunotherapeutic strategies that elicit protective cellular responses to HIV.
Acknowledgments
This work was supported by NIH R01 AI076066. K.R. Demers and M.A. Reuter were supported by NIH T32 AI007632.
Footnotes
The authors have no conflicts of interest to declare.
References
- 1.UNAIDS. Report on the Global AIDS Epdiemic. Geneva: UNAIDS; 2010. [Google Scholar]
- 2.Gilbert PB, et al. HIV-1 virologic and immunologic progression and initiation of antiretroviral therapy among HIV-1-infected subjects in a trial of the efficacy of recombinant glycoprotein 120 vaccine. J Infect Dis. 2005;192:974–983. doi: 10.1086/432734. [DOI] [PubMed] [Google Scholar]
- 3.Pitisuttithum P, et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis. 2006;194:1661–1671. doi: 10.1086/508748. [DOI] [PubMed] [Google Scholar]
- 4.Buchbinder SP, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet. 2008;372:1881–1893. doi: 10.1016/S0140-6736(08)61591-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rerks-Ngarm S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;361:2209–2220. doi: 10.1056/NEJMoa0908492. [DOI] [PubMed] [Google Scholar]
- 6.Montefiori DC, et al. Magnitude and breadth of the neutralizing antibody response in the RV144 and Vax003 HIV-1 vaccine efficacy trials. J Infect Dis. 2012;206:431–441. doi: 10.1093/infdis/jis367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Haynes BF, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 2012;366:1275–1286. doi: 10.1056/NEJMoa1113425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.McMichael AJ, Haynes BF. Lessons learned from HIV-1 vaccine trials: new priorities and directions. Nat Immunol. 2012;13:423–427. doi: 10.1038/ni.2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.McElrath MJ, et al. HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis. Lancet. 2008;372:1894–1905. doi: 10.1016/S0140-6736(08)61592-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Koup RA, et al. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol. 1994;68:4650–4655. doi: 10.1128/jvi.68.7.4650-4655.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol. 1994;68:6103–6110. doi: 10.1128/jvi.68.9.6103-6110.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Borrow P, et al. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nat Med. 1997;3:205–211. doi: 10.1038/nm0297-205. [DOI] [PubMed] [Google Scholar]
- 13.Goonetilleke N, et al. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. J Exp Med. 2009;206:1253–1272. doi: 10.1084/jem.20090365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Salazar-Gonzalez JF, et al. Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection. J Exp Med. 2009;206:1273–1289. doi: 10.1084/jem.20090378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Carrington M, O’Brien SJ. The influence of HLA genotype on AIDS. Annu Rev Med. 2003;54:535–551. doi: 10.1146/annurev.med.54.101601.152346. [DOI] [PubMed] [Google Scholar]
- 16.Dalmasso C, et al. Distinct genetic loci control plasma HIV-RNA and cellular HIV-DNA levels in HIV-1 infection: the ANRS Genome Wide Association 01 study. PLoS One. 2008;3:e3907. doi: 10.1371/journal.pone.0003907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Liu J, et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature. 2009;457:87–91. doi: 10.1038/nature07469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hansen SG, et al. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med. 2009;15:293–299. doi: 10.1038/nm.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hansen SG, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011;473:523–527. doi: 10.1038/nature10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Barouch DH, et al. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature. 2012;482:89–93. doi: 10.1038/nature10766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Catalina MD, Sullivan JL, Brody RM, Luzuriaga K. Phenotypic and functional heterogeneity of EBV epitope-specific CD8+ T cells. J Immunol. 2002;168:4184–4191. doi: 10.4049/jimmunol.168.8.4184. [DOI] [PubMed] [Google Scholar]
- 22.Gillespie GM, et al. Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors. J Virol. 2000;74:8140–8150. doi: 10.1128/jvi.74.17.8140-8150.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Appay V, et al. HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med. 2000;192:63–75. doi: 10.1084/jem.192.1.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hamann D, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med. 1997;186:1407–1418. doi: 10.1084/jem.186.9.1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Makedonas G, et al. Perforin and IL-2 upregulation define qualitative differences among highly functional virus-specific human CD8 T cells. PLoS Pathog. 2010;6:e1000798. doi: 10.1371/journal.ppat.1000798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147:258–267. [PubMed] [Google Scholar]
- 27.Groettrup M, et al. A role for the proteasome regulator PA28alpha in antigen presentation. Nature. 1996;381:166–168. doi: 10.1038/381166a0. [DOI] [PubMed] [Google Scholar]
- 28.Epperson DE, Arnold D, Spies T, Cresswell P, Pober JS, Johnson DR. Cytokines increase transporter in antigen processing-1 expression more rapidly than HLA class I expression in endothelial cells. J Immunol. 1992;149:3297–3301. [PubMed] [Google Scholar]
- 29.Cramer LA, Nelson SL, Klemsz MJ. Synergistic induction of the Tap-1 gene by IFN-gamma and lipopolysaccharide in macrophages is regulated by STAT1. J Immunol. 2000;165:3190–3197. doi: 10.4049/jimmunol.165.6.3190. [DOI] [PubMed] [Google Scholar]
- 30.Wallach D, Fellous M, Revel M. Preferential effect of gamma interferon on the synthesis of HLA antigens and their mRNAs in human cells. Nature. 1982;299:833–836. doi: 10.1038/299833a0. [DOI] [PubMed] [Google Scholar]
- 31.Johnson DR, Pober JS. Tumor necrosis factor and immune interferon synergistically increase transcription of HLA class I heavy- and light-chain genes in vascular endothelium. Proc Natl Acad Sci USA. 1990;87:5183–5187. doi: 10.1073/pnas.87.13.5183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tsujimoto M, Yip YK, Vilcek J. Interferon-gamma enhances expression of cellular receptors for tumor necrosis factor. J Immunol. 1986;136:2441–2444. [PubMed] [Google Scholar]
- 33.Xu X, Fu XY, Plate J, Chong AS. IFN-gamma induces cell growth inhibition by Fas-mediated apoptosis: requirement of STAT1 protein for up-regulation of Fas and FasL expression. Cancer Res. 1998;58:2832–2837. [PubMed] [Google Scholar]
- 34.Biswas P, et al. Interferon gamma induces the expression of human immunodeficiency virus in persistently infected promonocytic cells (U1) and redirects the production of virions to intracytoplasmic vacuoles in phorbol myristate acetate-differentiated U1 cells. J Exp Med. 1992;176:739–750. doi: 10.1084/jem.176.3.739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dinarello CA, Mier JW. Interleukins. Annu Rev Med. 1986;37:173–178. doi: 10.1146/annurev.me.37.020186.001133. [DOI] [PubMed] [Google Scholar]
- 36.Zimmerli SC, Harari A, Cellerai C, Vallelian F, Bart PA, Pantaleo G. HIV-1-specific IFN-gamma/IL-2-secreting CD8 T cells support CD4-independent proliferation of HIV-1-specific CD8 T cells. Proc Natl Acad Sci USA. 2005;102:7239–7244. doi: 10.1073/pnas.0502393102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258. doi: 10.1038/nri2274. [DOI] [PubMed] [Google Scholar]
- 38.Williams MA, Tyznik AJ, Bevan MJ. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature. 2006;441:890–893. doi: 10.1038/nature04790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Betts MR, et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107:4781–4789. doi: 10.1182/blood-2005-12-4818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Migueles SA, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol. 2002;3:1061–1068. doi: 10.1038/ni845. [DOI] [PubMed] [Google Scholar]
- 41.Davey RT, Jr, et al. Subcutaneous administration of interleukin-2 in human immunodeficiency virus type 1-infected persons. J Infect Dis. 1997;175:781–789. doi: 10.1086/513971. [DOI] [PubMed] [Google Scholar]
- 42.Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 1975;72:3666–3670. doi: 10.1073/pnas.72.9.3666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kull FC., Jr The TNF receptor in TNF-mediated cytotoxicity. Nat Immun Cell Growth Regul. 1988;7:254–265. [PubMed] [Google Scholar]
- 44.Lazdins JK, Grell M, Walker MR, Woods-Cook K, Scheurich P, Pfizenmaier K. Membrane tumor necrosis factor (TNF) induced cooperative signaling of TNFR60 and TNFR80 favors induction of cell death rather than virus production in HIV-infected T cells. J Exp Med. 1997;185:81–90. doi: 10.1084/jem.185.1.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3:745–756. doi: 10.1038/nri1184. [DOI] [PubMed] [Google Scholar]
- 46.Black RA, et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature. 1997;385:729–733. doi: 10.1038/385729a0. [DOI] [PubMed] [Google Scholar]
- 47.Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002;296:1634–1635. doi: 10.1126/science.1071924. [DOI] [PubMed] [Google Scholar]
- 48.Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10:45–65. doi: 10.1038/sj.cdd.4401189. [DOI] [PubMed] [Google Scholar]
- 49.Feldmann M, Brennan FM, Elliott MJ, Williams RO, Maini RN. TNF alpha is an effective therapeutic target for rheumatoid arthritis. Ann NY Acad Sci. 1995;766:272–278. doi: 10.1111/j.1749-6632.1995.tb26675.x. [DOI] [PubMed] [Google Scholar]
- 50.Scheurich P, Kronke M, Schluter C, Ucer U, Pfizenmaier K. Noncytocidal mechanisms of action of tumor necrosis factor-alpha on human tumor cells: enhancement of HLA gene expression synergistic with interferon-gamma. Immunobiology. 1986;172:291–300. doi: 10.1016/s0171-2985(86)80111-5. [DOI] [PubMed] [Google Scholar]
- 51.Duh EJ, Maury WJ, Folks TM, Fauci AS, Rabson AB. Tumor necrosis factor alpha activates human immunodeficiency virus type 1 through induction of nuclear factor binding to the NF-kappa B sites in the long terminal repeat. Proc Natl Acad Sci USA. 1989;86:5974–5978. doi: 10.1073/pnas.86.15.5974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Folks TM, et al. Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci USA. 1989;86:2365–2368. doi: 10.1073/pnas.86.7.2365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Harrer T, Jassoy C, Harrer E, Johnson RP, Walker BD. Induction of HIV-1 replication in a chronically infected T-cell line by cytotoxic T lymphocytes. J Acquir Immune Defic Syndr. 1993;6:865–871. [PubMed] [Google Scholar]
- 54.Callan MF. The evolution of antigen-specific CD8+ T cell responses after natural primary infection of humans with Epstein-Barr virus. Viral Immunol. 2003;16:3–16. doi: 10.1089/088282403763635401. [DOI] [PubMed] [Google Scholar]
- 55.Belz GT, Doherty PC. Virus-specific and bystander CD8+ T-cell proliferation in the acute and persistent phases of a gammaherpesvirus infection. J Virol. 2001;75:4435–4438. doi: 10.1128/JVI.75.9.4435-4438.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity. 1996;4:25–36. doi: 10.1016/s1074-7613(00)80295-2. [DOI] [PubMed] [Google Scholar]
- 57.Lechner F, et al. Analysis of successful immune responses in persons infected with hepatitis C virus. J Exp Med. 2000;191:1499–1512. doi: 10.1084/jem.191.9.1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Peters PJ, et al. Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J Exp Med. 1991;173:1099–1109. doi: 10.1084/jem.173.5.1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Shankar P, Xu Z, Lieberman J. Viral-specific cytotoxic T lymphocytes lyse human immunodeficiency virus-infected primary T lymphocytes by the granule exocytosis pathway. Blood. 1999;94:3084–3093. [PubMed] [Google Scholar]
- 60.Heusel JW, Wesselschmidt RL, Shresta S, Russell JH, Ley TJ. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell. 1994;76:977–987. doi: 10.1016/0092-8674(94)90376-x. [DOI] [PubMed] [Google Scholar]
- 61.Bots M, Medema JP. Granzymes at a glance. J Cell Sci. 2006;119:5011–5014. doi: 10.1242/jcs.03239. [DOI] [PubMed] [Google Scholar]
- 62.Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol. 2006;6:940–952. doi: 10.1038/nri1983. [DOI] [PubMed] [Google Scholar]
- 63.Bolitho P, Voskoboinik I, Trapani JA, Smyth MJ. Apoptosis induced by the lymphocyte effector molecule perforin. Curr Opin Immunol. 2007;19:339–347. doi: 10.1016/j.coi.2007.04.007. [DOI] [PubMed] [Google Scholar]
- 64.Trambas CM, Griffiths GM. Delivering the kiss of death. Nat Immunol. 2003;4:399–403. doi: 10.1038/ni0503-399. [DOI] [PubMed] [Google Scholar]
- 65.Rouvier E, Luciani MF, Golstein P. Fas involvement in Ca(2+)-independent T cell-mediated cytotoxicity. J Exp Med. 1993;177:195–200. doi: 10.1084/jem.177.1.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kischkel FC, et al. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995;14:5579–5588. doi: 10.1002/j.1460-2075.1995.tb00245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.He JS, Ostergaard HL. CTLs contain and use intracellular stores of FasL distinct from cytolytic granules. J Immunol. 2007;179:2339–2348. doi: 10.4049/jimmunol.179.4.2339. [DOI] [PubMed] [Google Scholar]
- 68.LAOR, et al. Membrane-bound Fas ligand only is essential for Fas-induced apoptosis. Nature. 2009;461:659–663. doi: 10.1038/nature08402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wajant H, Pfizenmaier K, Scheurich P. Non-apoptotic Fas signaling. Cytokine Growth Factor Rev. 2003;14:53–66. doi: 10.1016/s1359-6101(02)00072-2. [DOI] [PubMed] [Google Scholar]
- 70.Rollins BJ. Chemokines. Blood. 1997;90:909–928. [PubMed] [Google Scholar]
- 71.Catalfamo M, et al. Human CD8+ T cells store RANTES in a unique secretory compartment and release it rapidly after TcR stimulation. Immunity. 2004;20:219–230. doi: 10.1016/s1074-7613(04)00027-5. [DOI] [PubMed] [Google Scholar]
- 72.Wagner L, et al. Beta-chemokines are released from HIV-1-specific cytolytic T-cell granules complexed to proteoglycans. Nature. 1998;391:908–911. doi: 10.1038/36129. [DOI] [PubMed] [Google Scholar]
- 73.Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science. 1995;270:1811–1815. doi: 10.1126/science.270.5243.1811. [DOI] [PubMed] [Google Scholar]
- 74.Copeland KF. The role of CD8+ T cell soluble factors in human immunodeficiency virus infection. Curr Med Chem. 2002;9:1781–1790. doi: 10.2174/0929867023369006. [DOI] [PubMed] [Google Scholar]
- 75.Schmidtmayerova H, Sherry B, Bukrinsky M. Chemokines and HIV replication. Nature. 1996;382:767. doi: 10.1038/382767a0. [DOI] [PubMed] [Google Scholar]
- 76.Moriuchi H, Moriuchi M, Combadiere C, Murphy PM, Fauci AS. CD8+ T-cell-derived soluble factor(s), but not beta-chemokines RANTES, MIP-1 alpha, and MIP-1 beta, suppress HIV-1 replication in monocyte/macrophages. Proc Natl Acad Sci USA. 1996;93:15341–15345. doi: 10.1073/pnas.93.26.15341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Dragic T, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381:667–673. doi: 10.1038/381667a0. [DOI] [PubMed] [Google Scholar]
- 78.Trkola A, et al. The CC-chemokine RANTES increases the attachment of human immunodeficiency virus type 1 to target cells via glycosaminoglycans and also activates a signal transduction pathway that enhances viral infectivity. J Virol. 1999;73:6370–6379. doi: 10.1128/jvi.73.8.6370-6379.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Saha K, Bentsman G, Chess L, Volsky DJ. Endogenous production of beta-chemokines by CD4+, but not CD8+, T-cell clones correlates with the clinical state of human immunodeficiency virus type 1 (HIV-1)-infected individuals and may be responsible for blocking infection with non-syncytium-inducing HIV-1 in vitro. J Virol. 1998;72:876–881. doi: 10.1128/jvi.72.1.876-881.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Mackewicz CE, Barker E, Greco G, Reyes-Teran G, Levy JA. Do beta-chemokines have clinical relevance in HIV infection? J Clin Invest. 1997;100:921–930. doi: 10.1172/JCI119608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Walker CM, Moody DJ, Stites DP, Levy JA. CD8+ lymphocytes can control HIV infection in vitro by suppressing virus replication. Science. 1986;234:1563–1566. doi: 10.1126/science.2431484. [DOI] [PubMed] [Google Scholar]
- 82.Walker CM, Levy JA. A diffusible lymphokine produced by CD8+ T lymphocytes suppresses HIV replication. Immunology. 1989;66:628–630. [PMC free article] [PubMed] [Google Scholar]
- 83.Mackewicz CE, Ortega H, Levy JA. Effect of cytokines on HIV replication in CD4+ lymphocytes: lack of identity with the CD8+ cell antiviral factor. Cell Immunol. 1994;153:329–343. doi: 10.1006/cimm.1994.1032. [DOI] [PubMed] [Google Scholar]
- 84.Leith JG, Copeland KF, McKay PJ, Richards CD, Rosenthal KL. CD8+ T-cell-mediated suppression of HIV-1 long terminal repeat-driven gene expression is not modulated by the CC chemokines RANTES, macrophage inflammatory protein (MIP)-1 alpha and MIP-1 beta. AIDS. 1997;11:575–580. doi: 10.1097/00002030-199705000-00004. [DOI] [PubMed] [Google Scholar]
- 85.Rubbert A, et al. Multifactorial nature of noncytolytic CD8+ T cell-mediated suppression of HIV replication: beta-chemokine-dependent and -independent effects. AIDS Res Hum Retroviruses. 1997;13:63–69. doi: 10.1089/aid.1997.13.63. [DOI] [PubMed] [Google Scholar]
- 86.Vella C, Daniels RS. CD8+ T-cell-mediated non-cytolytic suppression of human immuno-deficiency viruses. Curr Drug Targets Infect Disord. 2003;3:97–113. doi: 10.2174/1568005033481196. [DOI] [PubMed] [Google Scholar]
- 87.Chang TL, Francois F, Mosoian A, Klotman ME. CAF-mediated human immunodeficiency virus (HIV) type 1 transcriptional inhibition is distinct from alpha-defensin-1 HIV inhibition. J Virol. 2003;77:6777–6784. doi: 10.1128/JVI.77.12.6777-6784.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chang TL, Mosoian A, Pine R, Klotman ME, Moore JP. A soluble factor(s) secreted from CD8(+) T lymphocytes inhibits human immunodeficiency virus type 1 replication through STAT1 activation. J Virol. 2002;76:569–581. doi: 10.1128/JVI.76.2.569-581.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Copeland KF, Leith JG, McKay PJ, Rosenthal KL. CD8+ T cell supernatants of HIV type 1-infected individuals have opposite effects on long terminal repeat-mediated transcription in T cells and monocytes. AIDS Res Hum Retroviruses. 1997;13:71–77. doi: 10.1089/aid.1997.13.71. [DOI] [PubMed] [Google Scholar]
- 90.Mackewicz CE, Patterson BK, Lee SA, Levy JA. CD8(+) cell noncytotoxic anti-human immunodeficiency virus response inhibits expression of viral RNA but not reverse transcription or provirus integration. J Gen Virol. 2000;81:1261–1264. doi: 10.1099/0022-1317-81-5-1261. [DOI] [PubMed] [Google Scholar]
- 91.Le Borgne S, Fevrier M, Callebaut C, Lee SP, Riviere Y. CD8(+)-Cell antiviral factor activity is not restricted to human immunodeficiency virus (HIV)-specific T cells and can block HIV replication after initiation of reverse transcription. J Virol. 2000;74:4456–4464. doi: 10.1128/jvi.74.10.4456-4464.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wong JK, et al. In vivo CD8+ T-cell suppression of siv viremia is not mediated by CTL clearance of productively infected cells. PLoS Pathog. 2010;6:e1000748. doi: 10.1371/journal.ppat.1000748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Klatt NR, et al. CD8+ lymphocytes control viral replication in SIVmac239-infected rhesus macaques without decreasing the lifespan of productively infected cells. PLoS Pathog. 2010;6:e1000747. doi: 10.1371/journal.ppat.1000747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Evans DT, et al. Virus-specific cytotoxic T-lymphocyte responses select for aminoacid variation in simian immunodeficiency virus Env and Nef. Nat Med. 1999;5:1270–1276. doi: 10.1038/15224. [DOI] [PubMed] [Google Scholar]
- 95.Allen TM, et al. Tat-specific cytotoxic T lymphocytes select for SIV escape variants during resolution of primary viraemia. Nature. 2000;407:386–390. doi: 10.1038/35030124. [DOI] [PubMed] [Google Scholar]
- 96.Goulder PJ, et al. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nat Med. 1997;3:212–217. doi: 10.1038/nm0297-212. [DOI] [PubMed] [Google Scholar]
- 97.Yant LJ, et al. The high-frequency major histocompatibility complex class I allele Mamu-B*17 is associated with control of simian immunodeficiency virus SIVmac239 replication. J Virol. 2006;80:5074–5077. doi: 10.1128/JVI.80.10.5074-5077.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Loffredo JT, et al. Mamu-B*08-positive macaques control simian immunodeficiency virus replication. J Virol. 2007;81:8827–8832. doi: 10.1128/JVI.00895-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jin X, et al. Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J Exp Med. 1999;189:991–998. doi: 10.1084/jem.189.6.991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Schmitz JE, et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science. 1999;283:857–860. doi: 10.1126/science.283.5403.857. [DOI] [PubMed] [Google Scholar]
- 101.Walker BD, et al. HIV-specific cytotoxic T lymphocytes in seropositive individuals. Nature. 1987;328:345–348. doi: 10.1038/328345a0. [DOI] [PubMed] [Google Scholar]
- 102.Ogg GS, et al. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science. 1998;279:2103–2106. doi: 10.1126/science.279.5359.2103. [DOI] [PubMed] [Google Scholar]
- 103.Addo MM, et al. Comprehensive epitope analysis of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome demonstrate broadly directed responses, but no correlation to viral load. J Virol. 2003;77:2081–2092. doi: 10.1128/JVI.77.3.2081-2092.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gea-Banacloche JC, et al. Maintenance of large numbers of virus-specific CD8+ T cells in HIV-infected progressors and long-term nonprogressors. J Immunol. 2000;165:1082–1092. doi: 10.4049/jimmunol.165.2.1082. [DOI] [PubMed] [Google Scholar]
- 105.Betts MR, et al. Analysis of total human immunodeficiency virus (HIV)-specific CD4(+) and CD8(+) T-cell responses: relationship to viral load in untreated HIV infection. J Virol. 2001;75:11983–11991. doi: 10.1128/JVI.75.24.11983-11991.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zajac AJ, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188:2205–2213. doi: 10.1084/jem.188.12.2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kalams SA, Walker BD. The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J Exp Med. 1998;188:2199–2204. doi: 10.1084/jem.188.12.2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Goulder PJ, et al. Functionally inert HIV-specific cytotoxic T lymphocytes do not play a major role in chronically infected adults and children. J Exp Med. 2000;192:1819–1832. doi: 10.1084/jem.192.12.1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yamamoto T, et al. Surface expression patterns of negative regulatory molecules identify determinants of virus-specific CD8+ T-cell exhaustion in HIV infection. Blood. 2011;117:4805–4815. doi: 10.1182/blood-2010-11-317297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Blackburn SD, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10:29–37. doi: 10.1038/ni.1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Trautmann L, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–1202. doi: 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
- 112.Petrovas C, et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J Exp Med. 2006;203:2281–2292. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Day CL, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–354. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
- 114.Jassoy C, et al. Human immunodeficiency virus type 1-specific cytotoxic T lymphocytes release gamma interferon, tumor necrosis factor alpha (TNF-alpha), and TNF-beta when they encounter their target antigens. J Virol. 1993;67:2844–2852. doi: 10.1128/jvi.67.5.2844-2852.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Morris AG, Lin YL, Askonas BA. Immune interferon release when a cloned cytotoxic T-cell line meets its correct influenza-infected target cell. Nature. 1982;295:150–152. doi: 10.1038/295150a0. [DOI] [PubMed] [Google Scholar]
- 116.Chen H, et al. Differential neutralization of human immunodeficiency virus (HIV) replication in autologous CD4 T cells by HIV-specific cytotoxic T lymphocytes. J Virol. 2009;83:3138–3149. doi: 10.1128/JVI.02073-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Rolland M, et al. Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial. Nat Med. 2011;17:366–371. doi: 10.1038/nm.2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yang H, et al. Antiviral inhibitory capacity of CD8+ T cells predicts the rate of CD4+ T-cell decline in HIV-1 infection. J Infect Dis. 2012;206:552–561. doi: 10.1093/infdis/jis379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kristensen NN, Christensen JP, Thomsen AR. High numbers of IL-2-producing CD8+ T cells during viral infection: correlation with stable memory development. J Gen Virol. 2002;83:2123–2133. doi: 10.1099/0022-1317-83-9-2123. [DOI] [PubMed] [Google Scholar]
- 120.Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol. 2003;77:4911–4927. doi: 10.1128/JVI.77.8.4911-4927.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sandberg JK, Fast NM, Nixon DF. Functional heterogeneity of cytokines and cytolytic effector molecules in human CD8+ T lymphocytes. J Immunol. 2001;167:181–187. doi: 10.4049/jimmunol.167.1.181. [DOI] [PubMed] [Google Scholar]
- 122.Perfetto SP, Chattopadhyay PK, Roederer M. Seventeen-colour flow cytometry: unravelling the immune system. Nat Rev Immunol. 2004;4:648–655. doi: 10.1038/nri1416. [DOI] [PubMed] [Google Scholar]
- 123.De Rosa SC, et al. Vaccination in humans generates broad T cell cytokine responses. J Immunol. 2004;173:5372–5380. doi: 10.4049/jimmunol.173.9.5372. [DOI] [PubMed] [Google Scholar]
- 124.Darrah PA, et al. IL-10 production differentially influences the magnitude, quality, and protective capacity of Th1 responses depending on the vaccine platform. J Exp Med. 2010;207:1421–1433. doi: 10.1084/jem.20092532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Darrah PA, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med. 2007;13:843–850. doi: 10.1038/nm1592. [DOI] [PubMed] [Google Scholar]
- 126.Emu B, et al. HLA class I-restricted T-cell responses may contribute to the control of human immunodeficiency virus infection, but such responses are not always necessary for long-term virus control. J Virol. 2008;82:5398–5407. doi: 10.1128/JVI.02176-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Rehr M, et al. Emergence of polyfunctional CD8+ T cells after prolonged suppression of human immunodeficiency virus replication by antiretroviral therapy. J Virol. 2008;82:3391–3404. doi: 10.1128/JVI.02383-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ortiz GM, et al. Structured antiretroviral treatment interruptions in chronically HIV-1-infected subjects. Proc Natl Acad Sci USA. 2001;98:13288–13293. doi: 10.1073/pnas.221452198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Davey RT, Jr, et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc Natl Acad Sci USA. 1999;96:15109–15114. doi: 10.1073/pnas.96.26.15109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Precopio ML, et al. Immunization with vaccinia virus induces polyfunctional and phenotypically distinctive CD8(+) T cell responses. J Exp Med. 2007;204:1405–1416. doi: 10.1084/jem.20062363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ferre AL, et al. Mucosal immune responses to HIV-1 in elite controllers: a potential correlate of immune control. Blood. 2009;113:3978–3989. doi: 10.1182/blood-2008-10-182709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zhang D, et al. Most antiviral CD8 T cells during chronic viral infection do not express high levels of perforin and are not directly cytotoxic. Blood. 2003;101:226–235. doi: 10.1182/blood-2002-03-0791. [DOI] [PubMed] [Google Scholar]
- 133.Andersson J, et al. Perforin is not co-expressed with granzyme A within cytotoxic granules in CD8 T lymphocytes present in lymphoid tissue during chronic HIV infection. AIDS. 1999;13:1295–1303. doi: 10.1097/00002030-199907300-00005. [DOI] [PubMed] [Google Scholar]
- 134.Doherty PC. The numbers game for virus-specific CD8+ T cells. Science. 1998;280:227. doi: 10.1126/science.280.5361.227. [DOI] [PubMed] [Google Scholar]
- 135.Betts MR, et al. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods. 2003;281:65–78. doi: 10.1016/s0022-1759(03)00265-5. [DOI] [PubMed] [Google Scholar]
- 136.Freel SA, et al. Initial HIV-1 antigen-specific CD8+ T cells in acute HIV-1 infection inhibit transmitted/founder virus replication. J Virol. 2012;86:6835–6846. doi: 10.1128/JVI.00437-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ferrari G, et al. Relationship between functional profile of HIV-1 specific CD8 T cells and epitope variability with the selection of escape mutants in acute HIV-1 infection. PLoS Pathog. 2011;7:e1001273. doi: 10.1371/journal.ppat.1001273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Freel SA, et al. Phenotypic and functional profile of HIV-inhibitory CD8 T cells elicited by natural infection and heterologous prime/boost vaccination. J Virol. 2010;84:4998–5006. doi: 10.1128/JVI.00138-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hersperger AR, Makedonas G, Betts MR. Flow cytometric detection of perforin upregulation in human CD8 T cells. Cytometry A. 2008;73:1050–1057. doi: 10.1002/cyto.a.20596. [DOI] [PubMed] [Google Scholar]
- 140.Wolint P, Betts MR, Koup RA, Oxenius A. Immediate cytotoxicity but not degranulation distinguishes effector and memory subsets of CD8+ T cells. J Exp Med. 2004;199:925–936. doi: 10.1084/jem.20031799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Makedonas G, et al. Rapid up-regulation and granule-independent transport of perforin to the immunological synapse define a novel mechanism of antigen-specific CD8+ T cell cytotoxic activity. J Immunol. 2009;182:5560–5569. doi: 10.4049/jimmunol.0803945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hersperger AR, et al. Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS Pathog. 2010;6:e1000917. doi: 10.1371/journal.ppat.1000917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Killian MS, Johnson C, Teque F, Fujimura S, Levy JA. Natural suppression of human immunodeficiency virus type 1 replication is mediated by transitional memory CD8+ T cells. J Virol. 2011;85:1696–1705. doi: 10.1128/JVI.01120-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Streeck H, et al. Human immunodeficiency virus type 1-specific CD8+ T-cell responses during primary infection are major determinants of the viral set point and loss of CD4+ T cells. J Virol. 2009;83:7641–7648. doi: 10.1128/JVI.00182-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Goepfert PA, et al. Transmission of HIV-1 Gag immune escape mutations is associated with reduced viral load in linked recipients. J Exp Med. 2008;205:1009–1017. doi: 10.1084/jem.20072457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wang W, Qiu C, Wang Y, Zhang X, Xu J. Development of skewed functionality of HIV-1-specific cytotoxic CD8(+) T cells from primary to early chronic phase of HIV infection. PLoS One. 2012;7:e44983. doi: 10.1371/journal.pone.0044983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Streeck H, et al. Antigen load and viral sequence diversification determine the functional profile of HIV-1-specific CD8+ T cells. PLoS Med. 2008;5:e100. doi: 10.1371/journal.pmed.0050100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Andersson J, et al. Low levels of perforin expression in CD8+ T lymphocyte granules in lymphoid tissue during acute human immunodeficiency virus type 1 infection. J Infect Dis. 2002;185:1355–1358. doi: 10.1086/340124. [DOI] [PubMed] [Google Scholar]
- 149.Appay V, et al. Dynamics of T cell responses in HIV infection. J Immunol. 2002;168:3660–3666. doi: 10.4049/jimmunol.168.7.3660. [DOI] [PubMed] [Google Scholar]
- 150.Cellerai C, et al. Early and prolonged antiretroviral therapy is associated with an HIV-1-specific T-cell profile comparable to that of long-term non-progressors. PLoS One. 2011;6:e18164. doi: 10.1371/journal.pone.0018164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Critchfield JW, et al. Magnitude and complexity of rectal mucosa HIV-1-specific CD8+ T-cell responses during chronic infection reflect clinical status. PLoS One. 2008;3:e3577. doi: 10.1371/journal.pone.0003577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Shacklett BL, et al. Abundant expression of granzyme A, but not perforin, in granules of CD8+ T cells in GALT: implications for immune control of HIV-1 infection. J Immunol. 2004;173:641–648. doi: 10.4049/jimmunol.173.1.641. [DOI] [PubMed] [Google Scholar]
- 153.Altfeld M, et al. Expansion of pre-existing, lymph node-localized CD8+ T cells during supervised treatment interruptions in chronic HIV-1 infection. J Clin Invest. 2002;109:837–843. doi: 10.1172/JCI14789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Connick E, et al. CTL fail to accumulate at sites of HIV-1 replication in lymphoid tissue. J Immunol. 2007;178:6975–6983. doi: 10.4049/jimmunol.178.11.6975. [DOI] [PubMed] [Google Scholar]
- 155.Folkvord JM, Armon C, Connick E. Lymphoid follicles are sites of heightened human immunodeficiency virus type 1 (HIV-1) replication and reduced antiretroviral effector mechanisms. AIDS Res Hum Retroviruses. 2005;21:363–370. doi: 10.1089/aid.2005.21.363. [DOI] [PubMed] [Google Scholar]
- 156.Perreau M, et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med. 2013;210:143–156. doi: 10.1084/jem.20121932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Petrovas C, et al. CD4 T follicular helper cell dynamics during SIV infection. J Clin Invest. 2012;122:3281–3294. doi: 10.1172/JCI63039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Lindqvist M, et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest. 2012;122:3271–3280. doi: 10.1172/JCI64314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Saez-Cirion A, et al. Heterogeneity in HIV suppression by CD8 T cells from HIV controllers: association with Gag-specific CD8 T cell responses. J Immunol. 2009;182:7828–7837. doi: 10.4049/jimmunol.0803928. [DOI] [PubMed] [Google Scholar]
- 160.Migueles SA, et al. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity. 2008;29:1009–1021. doi: 10.1016/j.immuni.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Miller SA, Weinmann AS. An essential interaction between T-box proteins and histone-modifying enzymes. Epigenetics. 2009;4:85–88. doi: 10.4161/epi.4.2.8111. [DOI] [PubMed] [Google Scholar]
- 162.Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell. 2000;100:655–669. doi: 10.1016/s0092-8674(00)80702-3. [DOI] [PubMed] [Google Scholar]
- 163.Joshi NS, 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]
- 164.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]
- 165.Sullivan BM, Juedes A, Szabo SJ, von Herrath M, Glimcher LH. Antigen-driven effector CD8 T cell function regulated by T-bet. Proc Natl Acad Sci USA. 2003;100:15818–15823. doi: 10.1073/pnas.2636938100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Jenner RG, et al. The transcription factors T-bet and GATA-3 control alternative pathways of T-cell differentiation through a shared set of target genes. Proc Natl Acad Sci USA. 2009;106:17876–17881. doi: 10.1073/pnas.0909357106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kao C, et al. Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nat Immunol. 2011;12:663–671. doi: 10.1038/ni.2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.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]
- 169.Pearce EL, et al. Control of effector CD8+ T cell function by the transcription factor Eomesodermin. Science. 2003;302:1041–1043. doi: 10.1126/science.1090148. [DOI] [PubMed] [Google Scholar]
- 170.Intlekofer AM, et al. Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet and eomesodermin. Science. 2008;321:408–411. doi: 10.1126/science.1159806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Intlekofer AM, et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat Immunol. 2005;6:1236–1244. doi: 10.1038/ni1268. [DOI] [PubMed] [Google Scholar]
- 172.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]
- 173.Banerjee A, et al. Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. J Immunol. 2010;185:4988–4992. doi: 10.4049/jimmunol.1002042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Joshi NS, Cui W, Dominguez CX, Chen JH, Hand TW, Kaech SM. Increased numbers of preexisting memory CD8 T cells and decreased T-bet expression can restrain terminal differentiation of secondary effector and memory CD8 T cells. J Immunol. 2011;187:4068–4076. doi: 10.4049/jimmunol.1002145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Taniuchi I, et al. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell. 2002;111:621–633. doi: 10.1016/s0092-8674(02)01111-x. [DOI] [PubMed] [Google Scholar]
- 176.Cruz-Guilloty F, et al. Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs. J Exp Med. 2009;206:51–59. doi: 10.1084/jem.20081242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Shin H, et al. A role for the transcriptional repressor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Immunity. 2009;31:309–320. doi: 10.1016/j.immuni.2009.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Rutishauser RL, 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]
- 179.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]
- 180.Ichii H, et al. Role for Bcl-6 in the generation and maintenance of memory CD8+ T cells. Nat Immunol. 2002;3:558–563. doi: 10.1038/ni802. [DOI] [PubMed] [Google Scholar]
- 181.Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ, Rao A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity. 2010;32:79–90. doi: 10.1016/j.immuni.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Cui W, Liu Y, Weinstein JS, Craft J, Kaech SM. An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity. 2011;35:792–805. doi: 10.1016/j.immuni.2011.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Yu D, et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity. 2009;31:457–468. doi: 10.1016/j.immuni.2009.07.002. [DOI] [PubMed] [Google Scholar]
- 184.Oestreich KJ, Huang AC, Weinmann AS. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate Th1 gene expression patterns. J Exp Med. 2011;208:1001–1013. doi: 10.1084/jem.20102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Oestreich KJ, Mohn SE, Weinmann AS. Molecular mechanisms that control the expression and activity of Bcl-6 in TH1 cells to regulate flexibility with a TFH-like gene profile. Nat Immunol. 2012;13:405–411. doi: 10.1038/ni.2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Wherry EJ, et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 2007;27:670–684. doi: 10.1016/j.immuni.2007.09.006. [DOI] [PubMed] [Google Scholar]
- 187.Paley MA, et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science. 2012;338:1220–1225. doi: 10.1126/science.1229620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.McLane LM, et al. Differential localization of T-bet and Eomes in CD8 T-cell memory populations. J Immunol. 2013 doi: 10.4049/jimmunol.1201556. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hersperger AR, et al. Increased HIV-specific CD8+ T-cell cytotoxic potential in HIV elite controllers is associated with T-bet expression. Blood. 2011;117:3799–3808. doi: 10.1182/blood-2010-12-322727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Ribeiro-dos-Santos P, et al. Chronic HIV infection affects the expression of the 2 transcription factors required for CD8 T-cell differentiation into cytolytic effectors. Blood. 2012;119:4928–4938. doi: 10.1182/blood-2011-12-395186. [DOI] [PubMed] [Google Scholar]
- 191.Cannarile MA, et al. Transcriptional regulator Id2 mediates CD8+ T cell immunity. Nat Immunol. 2006;7:1317–1325. doi: 10.1038/ni1403. [DOI] [PubMed] [Google Scholar]
- 192.Yang CY, et al. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat Immunol. 2011;12:1221–1229. doi: 10.1038/ni.2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Ji Y, et al. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells. Nat Immunol. 2011;12:1230–1237. doi: 10.1038/ni.2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Cho OH, et al. Notch regulates cytolytic effector function in CD8+ T cells. J Immunol. 2009;182:3380–3389. doi: 10.4049/jimmunol.0802598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Maekawa Y, et al. Notch2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nat Immunol. 2008;9:1140–1147. doi: 10.1038/ni.1649. [DOI] [PubMed] [Google Scholar]
- 196.Carter LL, Murphy KM. Lineage-specific requirement for signal transducer and activator of transcription (Stat)4 in interferon gamma production from CD4(+) versus CD8(+) T cells. J Exp Med. 1999;189:1355–1360. doi: 10.1084/jem.189.8.1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Xiao Z, Casey KA, Jameson SC, Curtsinger JM, Mescher MF. Programming for CD8 T cell memory development requires IL-12 or type I IFN. J Immunol. 2009;182:2786–2794. doi: 10.4049/jimmunol.0803484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Verdeil G, Puthier D, Nguyen C, Schmitt-Verhulst AM, Auphan-Anezin N. STAT5-mediated signals sustain a TCR-initiated gene expression program toward differentiation of CD8 T cell effectors. J Immunol. 2006;176:4834–4842. doi: 10.4049/jimmunol.176.8.4834. [DOI] [PubMed] [Google Scholar]
- 199.Lin JX, et al. Critical Role of STAT5 transcription factor tetramerization for cytokine responses and normal immune function. Immunity. 2012;36:586–599. doi: 10.1016/j.immuni.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]

