Abstract
HIV-specific cytotoxic T lymphocytes (CTL) are preferentially primed for apoptosis, and this may represent a viral escape mechanism. We hypothesized that HIV-infected individuals that control virus to undetectable levels without antiretroviral therapy (ART) (elite controllers [EC]) have the capacity to upregulate survival factors that allow them to resist apoptosis. To address this, we performed cross-sectional and longitudinal analysis of proapoptotic (cleaved caspase-3) and antiapoptotic (Bcl-2) markers of cytomegalovirus (CMV) and HIV-specific CD8 T cells in a cohort of HIV-infected subjects with various degrees of viral control on and off ART. We demonstrated that HIV-specific CTL from EC are more resistant to apoptosis than those with pharmacologic control (successfully treated patients [ST]), despite similar in vivo conditions. Longitudinal analysis of chronically infected persons starting ART revealed that the frequency of HIV-specific T cells prone to death decreased, suggesting that this phenotype is partially reversible even though it never achieves the levels present in EC. Elucidating the apoptotic factors contributing to the survival of CTL in EC is paramount to our development of effective HIV-1 vaccines. Furthermore, a better understanding of cellular markers that can be utilized to predict response durability in disease- or vaccine-elicited responses will advance the field.
INTRODUCTION
Without antiretroviral therapy (ART), the majority of HIV-infected individuals progress to AIDS. However, a small portion of those infected demonstrate evidence of immune control of the virus, namely, long-term nonprogressors (LTNP) and elite controllers (EC). These individuals can maintain low or undetectable viral loads without ART. Elucidating the mechanism of viral control in these unique patients remains an area of intense investigation. Despite scientific efforts over the past 2 decades, the design of an effective preventive vaccine for HIV still eludes us. Given the important role of CD8 T cell responses in viral control seen in nonhuman primate and human data (1, 2) and the fact that HLA class I alleles are associated with differences in disease progression (3–7), many current efforts are focused on defining an optimal CD8 T cell immune response to guide effective vaccine design. Unfortunately, studies have failed to consistently demonstrate clear associations of the breadth or magnitude of the cytotoxic T lymphocyte (CTL) response with the plasma viral load (pVL) (8–11). However, polyfunctional T cell responses, including the capacity to secrete cytokines, degranulate, and proliferate in response to antigen, correlate with clinical markers of disease progression (12–16). Maintenance of these types of responses also appears to be important, as patients identified and treated early generate and maintain these responses, while chronically infected patients with uncontrolled viremia and progressive disease lose these responses over time (17–21).
Although HIV-specific CTL appear to control HIV replication in most patients in acute infection and in EC or LTNP, we still do not understand why these responses are lost in the majority of patients in chronic infection (17, 22). CD8 T cells in chronic HIV infection succumb to exhaustion and cell death in an environment of uncontrolled viremia and nonspecific immune activation (23–25). Surface markers, including PD-1, CD160, and 2B4, have provided insights into predicting exhaustion and correlate with clinical parameters of disease progression (26). Similarly, vaccine design must incorporate the capacity to generate effective responses and maintain cell-mediated immunity over time or with subsequent boosting. The results of the RV 144 Thai Trial demonstrated modest protection overall that tended to be greatest in the first year but waned over time (27). This waning of vaccine efficacy highlights the importance of gaining a better understanding of the mechanisms dictating immune memory and the persistence of both antibodies and T cells. Furthermore, a better understanding of cellular markers that can be utilized to predict response durability in disease- or vaccine-elicited responses would advance the field.
Apoptosis occurs through two main pathways. The extrinsic pathway is mediated by surface death receptors, such as Fas/FasL. The intrinsic pathway is an intracellular process that can be initiated by a variety of mechanisms, including lack of growth factors or cytokines, that result in mitochondrial damage (28). Pro- and antiapoptotic members of the Bcl-2 family of proteins regulate the subsequent mitochondrial release of cytochrome c to induce apoptosis (29–31). Antiapoptotic members of the Bcl-2 family, such as Bcl-2 and Bcl-XL, negatively regulate the induction of the intrinsic apoptotic pathway (30). The intrinsic and extrinsic pathways converge on caspase-3, the effector caspase. Cleavage of caspase-3 results in a cascade of events that lead to programmed cell death (32). Thus, measurement of cleaved caspase-3 (CC-3) reflects cell death occurring by either the intrinsic or extrinsic pathway.
While most research on apoptosis and HIV has focused on the death of HIV-infected CD4 T cells, there is a growing body of literature investigating the death of CD8 T cells and how this may represent an escape mechanism for the virus. This statement is especially pertinent in light of the evidence that apoptosis of the total CD8 T cell population correlates with disease progression (23, 33, 34). Furthermore, analysis of total CD8 T cell apoptosis in LTNP has demonstrated that LTNP have fewer apoptotic CD8 T cells (35, 36). The importance of this phenomenon is more accurately reflected by studies of HIV-specific CTL. In a cross-sectional analysis of chronically infected patients, HIV-specific CD8 T cells were more susceptible to Fas-mediated cell death than cytomegalovirus (CMV)-specific CD8 T cells from the same patient (37) and had reduced levels of Bcl-2 and Bcl-XL (38). Furthermore, despite similar levels of Fas on CMV- and HIV-specific cells from the same patient, HIV-specific CTL were 3-fold more prone to apoptosis (37). In vitro studies also demonstrate that HIV-infected, activated macrophages (MΦ) can induce apoptosis in HIV-specific CTL, suggesting that while engaging with HIV-infected targets, CTL are at risk of being killed by the targets they are attempting to destroy (37). While the study of apoptosis of cytotoxic T cells suggests a putative mechanism that explains the loss of viral control, studies to date have been limited, as cohorts have not been divided by immune status, pVL, or receipt of ART. In addition, previous studies have not analyzed EC in whom survival of optimal CTL may explain why these patients demonstrate durable viral control. We hypothesize that the capacity for prolonged survival rather than exhaustion and death may be a unique and desirable quality of HIV-specific T cells. Thus, we sought to define the role of apoptosis resistance via upregulation of survival molecules, such as Bcl-2, in the maintenance of effective CTL responses in EC and to elucidate the signaling mechanisms involved in generating those responses. This capacity to resist apoptosis may be an important defining advantage of EC in their control of viremia. Furthermore, CTL with increased survival capacity or apoptosis resistance may be protected from the deleterious effects of immune activation, exhaustion, and death.
MATERIALS AND METHODS
Ethics statement.
Certification by the UAB IRB indicating their review and approval of this activity in accordance with the Common Rule and any other governing regulations has been issued and is on file with the Department of Health and Human Services. Written, informed consent, approved by the international review board, was obtained from each of the study subjects.
Study subjects and HLA typing.
Peripheral blood samples were collected from healthy donors and HIV-infected volunteers at University of Alabama at Birmingham (UAB) 1917 HIV and Alabama Vaccine Research Clinics. HLA typing was performed using the Micro SSP HLA typing system. Table 1 lists the clinical parameters of the patient cohort, which was divided into the following groups: elite controllers (EC), i.e., subjects maintaining plasma HIV-1 viral RNA loads (pVL) below the limit of detection by commercial assays (<50 copies/ml) without antiretroviral therapy (ART) for at least 4 years; successfully treated patients (ST), i.e., subjects with undetectable pVL on ART for a mean of 22 months (range, 10 to 47 months); viremic controllers (VC), i.e., subjects with detectable pVL which always remains below 2,000 copies/ml without ART; and progressors (P), i.e., subjects with higher pVL (>2,000 copies/ml) off ART. Viral blips of <200 copies/ml were acceptable in subjects with an undetectable viral load if subsequent viral load measurements were <50 copies/ml. Healthy controls were HIV-seronegative subjects (n = 9). HLA-specific analysis of CD8 T cells was restricted to HLA-A*2 and HLA-B*57. Table 2 shows detailed information on each patient, including viral load, receipt of antiretrovirals, CD4 T cell count, age, gender, HLA type, gamma interferon (IFN-γ) enzyme-linked immunosorbent spot assay (ELISpot) data, and frequency of HIV- and CMV-specific T cells as measured by tetramer.
Table 1.
Clinical characteristics of HIV-1-infected patient cohort
| Patient group | No. of patients | Plasma viral load (RNA copies/ml) | Absolute CD4 count [range (median no. of cells/ml)] |
|---|---|---|---|
| Aviremic | |||
| Elite controller | 3 | <50 | 850–1,750 (890) |
| Successfully treated on ARTa | 5 | <50 | 450–650 (573) |
| Viremic | |||
| Viremic controller | 4 | <50–955 | 400–650 (583) |
| Progressor | 11 | 2,000–80,000 | 250–650 (424) |
Note that these patients have undetectable VL on ART.
Table 2.
Clinical parameters of HIV-infected subject cohorta
| Patient group and identifier | Viral load (RNA copies/ml) | ART status | CD4 count (cells/ml) | Age | Gender | HLA type(s) | SL9 response |
KF11 response |
NV9 response (% tetramer+) | ||
|---|---|---|---|---|---|---|---|---|---|---|---|
| SFU/million cells | % tetramer+ | SFU/million cells | % tetramer+ | ||||||||
| EC 1655 | <50 | Naïve | 1,741 | 45 | F | A2, B57 | 90 | 1.07 | 70 | 1.76 | 1.4 |
| EC 5823 | <50 | Naïve | 890 | 51 | M | A2, B57 | 2,285 | 3.79 | 2,815 | 2.59 | 2.84 |
| EC 5915b | 178 | Naïve | 850 | 44 | M | A2 | 260 | 0.18 | NA | NA | ND |
| ST 4081 | <50 | On | 623 | 59 | M | A2 | 105 | 0.52 | NA | NA | 3.43 |
| ST 3814 | <50 | On | 576 | 60 | F | A2, B57 | 1,000 | 1.86 | 1,000 | 1.58 | 1.84 |
| ST 4447 | <50 | On | 458 | 52 | M | A2, B57 | 1,000 | 1.27 | 1,000 | 1.99 | ND |
| ST 4480 | <50 | On | 550 | 31 | M | B57 | NA | NA | 450 | 1.59 | NA |
| ST 4496 | <50 | On | 573 | 51 | F | A2 | 310 | 0.51 | NA | NA | ND |
| VC 2795c | <50 | Off | 404 | 48 | M | B57 | NA | NA | 4,265 | 4.12 | NA |
| VC 2976 | 453 | Naïve | 863 | 37 | F | B57 | NA | NA | 125 | 0.37 | NA |
| VC 4311 | 873 | Naïve | 524 | 47 | M | A2 | 337 | 1.56 | NA | NA | ND |
| VC 6351 | 955 | Naïve | 642 | 39 | M | B57 | NA | NA | 680 | 2.22 | NA |
| P 6410 | 1,728 | Naïve | 847 | 27 | F | A2, B57 | — | 0.88 | 715 | 1.17 | ND |
| P 4024 | 5,258 | Naïve | 289 | 34 | M | A2, B57 | 300 | 0.41 | 310 | 0.83 | ND |
| P 4447 | 12,768 | Naïve | 501 | 52 | M | A2, B57 | 1,320 | 1.27 | 2,650 | 1.87 | ND |
| P 4496 | 14,208 | Off | 260 | 51 | F | A2 | 310 | 1.42 | NA | NA | 0.88 |
| P 4081 | 41,681 | Off | 436 | 59 | M | A2 | 105 | 0.6 | NA | NA | 3.21 |
| P 6315 | 43,500 | Naïve | 571 | 39 | M | B57 | NA | NA | 680 | 2.02 | NA |
| P 2569 | 53,876 | Off | 406 | 42 | M | A2, B57 | — | 0.36 | 1,058 | 3.15 | 0.62 |
| P 4480 | 65,030 | Off | 424 | 31 | M | B57 | NA | NA | 450 | 3.11 | NA |
| P 4081 | 68,139 | Naïve | 408 | 59 | M | A2 | 105 | 0.44 | NA | NA | 3.24 |
| P 1581 | 79,286 | Naïve | 672 | 41 | M | A2, B57 | — | 1.4 | 330 | 1.93 | 1.72 |
| P 4480 | 264,120 | Off | 294 | 31 | M | B57 | NA | NA | 450 | 1.48 | NA |
SFU, spot-forming units; F, female; M, male; NA, data not available (individual did not have the HLA allele restricting the specific peptide tested); —, response not detected (<50 SFU/million cells); ND, not done.
Patient had viral blip of 178 copies/ml.
All prior viral load measurements ranged from 81 to 458 copies/ml.
Quantitation of HIV-1 RNA in plasma and absolute CD4+ T cell count.
Plasma HIV-1 RNA levels were measured using the Amplicor ultrasensitive HIV-1 monitor assay at UAB hospitals (version 1.5; Roche Diagnostics Systems). Peripheral blood CD4+ T cells were determined as described elsewhere (39). Patients are routinely seen at 3-month intervals, and CD4 counts and HIV viral load measurements are obtained at each clinic visit.
Antigens.
HLA-restricted HIV-specific responses were determined using the IFN-γ ELISpot and optimized 9- to 11-mer peptides (Los Alamos National Laboratory HIV Immunology Database). Peripheral blood mononuclear cells (PBMC) were stimulated using optimized HLA-restricted CD8 T cell epitopes, including HLA B*57-restricted Gag-p24 KF11 (KAFSPEVIPMF) and HLA-A*2-restricted Gag-p17 SL9 (SLYNTVATL) (10 μM), cytomegalovirus (CMV) pp65 NV9 (NLVPMVATV) (10 μmol/liter), or staphylococcal enterotoxin B (SEB) (1 μg/ml) (positive control). Peptides previously shown to elicit IFN-γ ELISPOT responses in the studied patients were chosen for further studies.
Ex vivo or in vitro analysis.
Cryopreserved PBMC were thawed, washed in complete RPMI medium (10% AB serum) (R-10 medium), and rested overnight at 37°C in a 5% CO2 incubator. Cells were counted using Trypan blue staining, and 1 × 106 PBMC were analyzed ex vivo or were cultured in 500 μl R-10 in short-term (6-h; ex vivo) and long-term (4-day; in vitro) cultures at 37°C and 5% CO2. The peptides HIV Gag-p24 KF11 and -p17 SL9 or CMV pp65 NV9 were used at a concentration of 10 μM for 4 days of peptide stimulation where indicated below.
Surface staining and ICCS.
PBMC from ex vivo and in vitro studies were stained with tetramer (allophycocyanin [APC] HLA-B*57-KF11, APC HLA-A*2-SL9, or PE HLA-A*2-CMV-NV9) (Beckman Coulter) and surface markers (Pacific blue anti-CD3, APC-Alexa Fluor 750 anti-CD4, peridinin chlorophyll protein [PerCp]-Cy5.5 anti-CD8, and phycoerythrin [PE]-Cy7 [PECy7] anti-CD45RO antibodies) (BD Biosciences). Intracellular cytokine staining (ICCS) was performed as described elsewhere (40–42) and was utilized for the detection of cleaved caspase-3 (CC-3) and Bcl-2. Briefly, cells were washed with phosphate-buffered saline (PBS) and permeabilized using Cytofix/Cytoperm (BD Biosciences). Intracellular staining was performed using fluorescein isothiocyanate (FITC)–anti-cleaved caspase-3 (Cell Signaling) and PE–anti-Bcl-2 (BD Biosciences) antibodies according to the manufacturer's protocol.
Flow cytometric analysis.
CompBeads (BD Biosciences) were used to establish the fluorescence compensation setting for multicolor flow cytometric analysis. A minimum of 5 × 105 CD3+ events were collected using an LSRII flow cytometer (Becton, Dickinson) and analyzed using FlowJo software version 8.5.2, as previously described (42). Since we were interested in studying apoptosis, we did not include a viability stain, as this could reduce the events of interest. Lymphocytes were gated on forward scatter area (FSC-A) versus side scatter area (SSC-A), and any cells that had low forward scatter with higher side scatter were excluded. Next, CD3+ CD8+ T cells were selected. From this population, total CD8 T cells or tetramer-specific T cells were analyzed for cleaved caspase-3 expression (see Fig. 1 for gating strategy). Bcl-2, which is expressed in naïve cells, decreases at the peak of the expansion phase and remains low in cells that undergo apoptosis (43). Memory cells typically upregulate Bcl-2 expression, a quality that aids in their survival (43). Thus, all Bcl-2 expression was analyzed from the CD45RO+ memory CD3+ CD8+ gate to exclude activation-induced Bcl-2 downregulation and allow comparison of the memory population (see Fig. 3A and Fig. 4A for gating strategies) (43).
Fig 1.
Frequencies of cleaved caspase-3+ CD8+ T cells in HIV-infected patients. (A) Representative results of indicated polychromatic flow cytometry gating scheme for measurement of cleaved caspase-3 levels on total and tetramer-specific CD8+ T cells are shown. (B) Percentages of CC-3hi CD8+ T cells in healthy controls compared to those in HIV-infected subjects. In both groups, total CD8 T cells are compared to virus tetramer-specific (HIV+ or CMV+) CD8 T cells. (C) Frequencies of CC-3hi total CD8 T cells in healthy, aviremic (elite controllers and successfully treated patients) and viremic (viremic controllers and progressors) subjects. (D) Frequencies of CC-3hi (tetramer-specific [CMV+ or HIV+]) CD8 T cells in HIV-aviremic and HIV-viremic subjects. CMV-sp, CMV specific. Note that the symbols for panel D (diamonds, progressors; squares, viremic controllers; triangles, successfully treated patients; circles, elite controllers) apply to both HIV-specific and CMV-specific responses. *, P < 0.05.
Fig 3.
Ex vivo expression of Bcl-2 in total and HIV-specific CD8 T cells. (A) Representative results of indicated flow cytometric gating scheme for measurement of Bcl-2 expression on total CD8+ T cells are shown. (B) Total CD45R0+ CD8 T cells were examined for expression of the prosurvival molecule Bcl-2. The frequencies of Bcl-2hi total CD8+ T cells in healthy controls versus HIV patients, including aviremic (elite controllers and successfully treated patients) and viremic (viremic controllers and progressors) subjects, are depicted. (C) HIV-specific (tetramer+) CD45R0+ CD8 T cells were examined for expression of the prosurvival molecule Bcl-2. Frequencies of Bcl-2hi HIV-specific CD8 T cells are plotted across the cohort. *, P < 0.05.
Fig 4.
Coexpression of Bcl-2 and CC-3 on HIV-specific CD8 T cells. (A) Representative results of indicated polychromatic flow cytometry gating scheme for measurement of cleaved caspase-3 and Bcl-2 expression on CD45RO+ tetramer+ CD8+ T cells are shown. Gating from lymphocytes to CD3+ CD8+ T cells is the same as depicted in Figure 1A. The frequencies of CC-3lo Bcl-2hi HIV-specific CD8 T cells (primed for survival) or CC-3hi Bcl-2lo HIV-specific CD8 T cells (prone to death) were determined for each subject. (B) Coexpression of CC-3 and Bcl-2 is plotted using the frequencies of CC-3lo Bcl-2hi HIV-specific CD8 T cells (primed for survival) for each subject across the cohort. *, P < 0.05. (C) Four patients were examined longitudinally before the initiation of ART and 1 year after the initiation of ART. Frequencies of CC-3lo Bcl-2hi HIV-specific CD8 T cells (gated on tetramer+ cells), cells primed for survival, are plotted with connecting solid lines to demonstrate the effect of ART. Dotted line connecting open symbols demonstrates the frequencies of CC-3lo Bcl-2hi CMV-specific CD8 T cells from one of the ART-treated subjects before and after ART treatment. (D) Frequencies of CC-3hi Bcl-2lo HIV-specific CD8 T cells (gated on tetramer+ cells), cells prone to death, are plotted with connecting solid lines to demonstrate ART effect. Dotted line connecting open symbols demonstrates the frequencies of CC-3hi Bcl-2lo CMV-specific CD8 T cells from one of the ART-treated subjects before and after ART treatment.
Statistics.
Using Prism software (version 5 GraphPad), statistical analysis included the nonparametric Mann-Whitney U test to compare differences between categorical variables. Spearman rank was used as the nonparametric test to compare the relationship between continuous variables. Statistically significant differences were defined as those with a P value of <0.05.
RESULTS
Ex vivo expression of cleaved caspase-3 in HIV-specific CTL.
To address the potential for apoptosis resistance as a mechanism of maintaining effective CD8 T cells in the rare HIV-infected subjects capable of immune control of HIV, we analyzed subjects who demonstrated various levels of HIV control with and without antiretroviral treatment (Tables 1 and 2). We performed an ex vivo flow cytometric analysis on PBMC, analyzing cleaved caspase-3 (CC-3), a marker of apoptosis. Caspase-3 is the terminal effector caspase for both intrinsic (mitochondrial-dependent) and extrinsic (death receptor ligand-dependent) apoptosis pathways. Thus, measurement of CC-3 accurately reflects the induction of apoptosis resulting from cytokine deficiencies and/or surface signaling via Fas and TRAIL. We performed tetramer staining using HLA-B*57 Gag KF11, HLA-A*2 Gag SL9, and HLA-A*2 CMV NV9 tetramers to allow analysis of HIV- and CMV-specific responses. For HIV-specific T cells, we limited our analysis to HLA-B*57 and HLA-A*2 Gag-restricted responses. Doing so allowed us to compare responses restricted by a favorable HLA (i.e., B*57) and a neutral HLA (i.e., A*2). The HLA of each patient from each patient group can be found in Table 2. Representative results of the gating strategy to measure CC-3 on total and HIV-specific CD8 T cells are shown (Fig. 1A), as well as a summary of the results for the entire cohort (Fig. 1B). Similar to prior studies, we found that total CD8 T cells from HIV-infected persons are more prone to apoptosis than those from healthy controls (P = 0.0001) and that HIV-specific CD8 T cells (tetramer positive [tetramer+]) are 2 to 3 times more prone to cell death than CMV-specific cells from the same persons (P = 0.02) (Fig. 1B). When examining apoptosis of total CD8 T cells, cells from aviremic HIV-infected persons (EC and ST) were similar to those from healthy controls (Fig. 1C). In contrast, the total CD8 T cells from viremic patients (VC and P) were more prone to apoptosis than those of healthy controls (P = 0.005 and P = 0.0001, respectively) (Fig. 1C). When comparing HIV-specific responses in these cohorts, we found no differences in cell death between viremic individuals (VC and P), despite viremic controllers maintaining pVL of <2,000 copies/ml without ART (Fig. 1D). This suggests that the mere presence of circulating virus, rather than the level of viremia, resulted in an increased frequency of cells prone to death. In contrast, we found significant differences in the frequency of HIV-specific T cells prone to death as measured by CC-3 in the aviremic group, with EC having the lowest frequency of cells prone to apoptosis (P = 0.048) (Fig. 1D). We also examined HIV-specific CD8 T cells targeting either HLA A*2 Gag SL9 or HLA B*57 Gag KF11 to determine if responses restricted by favorable HLA were more likely to survive under unfavorable conditions. The levels of CC-3 on HIV-specific CD8 T cells were indistinguishable between these two HLA phenotypes within each of the groups (data not shown).
Cleaved caspase-3 levels on HIV-specific CTL after stimulation.
We found significant differences in the ex vivo levels of CC-3 in HIV-specific (tetramer+) CD8 T cells between groups. In vivo, CD8 T cells from EC and ST have minimal virus exposure. We next sought to extend these studies to determine the effect of HIV peptide stimulation over time on cell death and survival. For these studies, we cultured PBMC from HIV-positive subjects for 4 days in the presence (stimulated) or absence (medium alone) of HIV peptides (KF11 or SL9). In addition, PBMC from healthy HIV-seronegative controls were cultured under similar conditions in the presence or absence of CMV peptide NV9. In our flow cytometric analysis of CC-3 under ex vivo conditions, we consistently found that the subset of HIV-specific CD8 T cells that had downregulated CD8 (CD8dim) were the most prone to cell death (as determined by increased levels of CC-3) (Fig. 2A and D). In fact, 80 to 90% of the CC-3hi HIV-specific CD8 T cells originated from the CD8dim subset of CTL (Fig. 2B and D). This was observed across all HIV subgroups. We therefore analyzed both populations (CD8hi and CD8dim T cells) for CC-3 levels within our viremic and aviremic cohorts in an ex vivo and a 4-day culture (medium alone or with simultaneous peptide stimulation) assay. Across all HIV subgroups, we observed lower levels of CC-3 in the CD8hi T cell group (Fig. 2A and C). Interestingly, peptide stimulation of PBMC over 4 days resulted in statistically higher levels of CC-3 in the CD8hi population in both viremic populations (Fig. 2B). Alternatively, we found that CD8dim cells were much more prone to death across the HIV cohort and under all conditions, as evidenced by higher expression of CC-3 (Fig. 2A and D). These cells demonstrated sensitivity to cell death in long-term culture, even in the absence of peptide stimulation (Fig. 2D, hatched versus white bars), that was present regardless of patient group. However, the magnitude of the cell death was remarkably different across the cohort. Despite overall higher expression of CC-3 among the CD8dim cells than in the CD8hi cells, EC consistently maintained the lowest levels of CC-3 within the CD8dim population across the cohort. This observation was noted under both of the 4-day culture conditions, where the expression of CC-3 by EC was found to be 4-fold lower than for viremic (VC and P) subjects and 2-fold lower than for ST subjects (Fig. 2D).
Fig 2.
Comparison of rates of cell death between ex vivo and long-term culture with and without peptide stimulation. (A) Representative results of indicated polychromatic flow cytometry gating scheme to measure the frequencies of CC-3hi in HIV-specific CD8+ T cells (gated on tetramer+ cells) in ex vivo studies (left) and after 4 days of in vitro stimulation with HIV peptides (right) are shown. CD8 T cell population is divided to demonstrate CD8hi versus CD8dim, the subpopulation with CD8 downregulation. (B) Frequencies of CC-3hi antigen-specific (Ag-sp) CD8hi + dim cells (gated on tetramer+ cells) across the cohort under different culture conditions. Antigen-specific responses for healthy individuals are to CMV (pp65-NV9 tetramer) and for HIV-infected individuals to HIV-1 (Gag-p17 SL9 and Gag-p24 KF11 tetramers). (C) Frequencies of CC-3hi antigen-specific CD8hi cells (gated on tetramer+ cells) across the cohort under different culture conditions. (D) Frequencies of CC-3hi antigen-specific CD8dim cells (gated on tetramer+ cells) across the cohort under different culture conditions. *, P < 0.05.
Ex vivo expression of Bcl-2 in total and HIV-specific CD8 T cells.
Since the expression of antiapoptotic molecules can affect the fate of the cell, we sought to determine the ex vivo levels of antiapoptotic Bcl-2 in cells derived from the same patient cohort. When examining Bcl-2 levels in total CD8 T cell populations, VC were different from healthy individuals and exhibited significantly higher levels than P (Fig. 3B). The levels of Bcl-2 on total CD8 T cells observed in healthy, EC, and ST patients were comparable. However, upon analysis of HIV-specific CD8 T cells (Gag tetramer+), we found significant upregulation of Bcl-2 in EC and ST compared to the levels in viremic subjects (Fig. 3C). Among HIV-specific CD8 T cells, EC exhibited approximately 2-fold higher levels of Bcl-2 than ST and more than 3-fold-higher levels than both viremic groups.
HIV-specific CTL from elite controllers have a survival advantage.
Analysis of CD8 T cells in EC indicates increased levels of antiapoptotic Bcl-2 and reduced levels of proapoptotic CC-3 compared to the levels in cells obtained from other HIV-infected groups. We next utilized a CC-3 and Bcl-2 colabeling strategy to provide additional insight as to the percentage of cells in each population that were more effectively primed for survival (CC-3lo Bcl-2hi) (Fig. 4A). We found significant differences between each group, demonstrating that EC and ST have fewer cells at risk of apoptosis than viremic subjects. Interestingly, when comparing subjects with immune control of virus (EC) to those with pharmacologic control of virus (ST) in an ex vivo analysis, HIV-specific CTL from the EC exhibited a survival advantage, demonstrating preservation of cells primed for survival (CC-3lo Bcl-2hi) (for EC versus ST, P = 0.038) (Fig. 4B). This survival phenotype was not preserved in viremic controllers when compared to progressors, despite their having some level of immune viral control. In contrast, EC maintained the lowest frequency of cells prone to death (CC-3hi Bcl-2lo) (data not shown).
Treatment partially reverses the susceptibility of HIV-specific CTL to apoptosis.
Since we found that the pattern of pro- and antiapoptotic molecule expression in ST patients more closely mimicked that of the EC group but never achieved the same levels, we extended these studies by performing a longitudinal analysis of four chronically infected persons at least 1 year prior to initiating ART and at 1 year after ART initiation, once viremia was suppressed to undetectable levels (Fig. 4C and D). We demonstrated that control of viremia with ART resulted in upregulation of Bcl-2 and downregulation of CC-3 in HIV-specific CTL, a pattern consistent with improved cell survival (Fig. 4C). In addition, the frequency of HIV-specific CD8 T cells prone to death (CC-3hi Bcl-2lo) declined dramatically with suppression of HIV (Fig. 4D, closed symbols), approaching the frequency of CMV-specific CD8 T cells with the same phenotype (Fig. 4D, open symbols). Thus, ART, while unable to reduce apoptotic cells to the levels seen in EC, was able to partially reverse the survival defect demonstrated in HIV-specific CTL.
Longitudinal data before and after the initiation of ART suggest that circulating virus, in part, contributes to enhanced cell death of HIV-specific CD8 T cells. However, the finding that VC with pVL of <2,000 copies/ml have frequencies of cells prone to apoptosis similar to the frequencies in patients with poorly controlled virus (Fig. 1D) challenges the notion that circulating virus is the primary driver of cell death. To better understand the extent of viremia driving cell death, we compared the frequencies of total (Fig. 5A) and HIV-specific (Fig. 5B) CC-3hi CD8 T cells to plasma viral loads and found that viral load had a positive correlation, albeit weak, with CD8 T cell CC-3 expression (R2 = 0.20 and P = 0.03 for total CD8 T cells and R2 = 0.29 and P = 0.001 for HIV-specific CD8 T cells). For a more stringent analysis, we excluded all subjects with an undetectable plasma viral load (pVL < 50), including elite controllers and successfully treated patients. This analysis suggests that viral load was a poor correlate of apoptosis (R2 = 0.0007 and P = 0.738) (Fig. 5C).
Fig 5.
Correlation of frequency of CC-3hi CD8 T cells with plasma viral load. (A) For each patient, the frequency of CC-3hi total CD8 T cells in ex vivo analysis is plotted versus pVL. (B) CC-3hi HIV-specific CD8 T cells are plotted versus pVL. (C) Aviremic subjects were excluded, and the analysis of CC-3hi HIV-specific CD8 T cells is plotted versus pVL in viremic subjects only. Spearman rank correlation was performed.
CMV-specific apoptosis in HIV-seronegative and -seropositive subjects.
While the focus of this work was on HIV-specific CD8 T cells, we also examined the susceptibility of CMV-specific CD8 T cells from both HIV-seronegative subjects and HIV-infected subjects. CMV-specific CD8 T cells from HIV-positive subjects are more prone to apoptosis than CMV-specific CD8 T cells from HIV-seronegative subjects (Fig. 1B, closed squares versus open squares). Within HIV-infected subjects, we observed a modest range of susceptibility of CMV-specific T cells to apoptosis, with cells from EC and ST being the least susceptible and cells from progressors being the most susceptible (Fig. 1D, gray symbols). Despite the range, all CMV-specific CD8 T cells had relatively low CC-3 expression, comparable to that of HIV-specific CD8 T cells from EC and ST (Fig. 1D). This is perhaps a reflection that CMV viremia is controlled better than HIV viremia. In long-term culture, CMV-specific T cells from healthy HIV-seronegative controls had low levels of apoptosis under ex vivo and 4-day culture conditions (Fig. 2B). Cell death increased with CMV peptide stimulation, but the frequencies of CC-3hi CD8+ cells were comparable between the CD8hi and CD8dim populations (Fig. 2C and D). Finally, we examined the coexpression of CC-3 and Bcl-2 by CMV-specific CD8 T cells from a progressor who was eventually treated with ART and was reanalyzed at a later time point when virus was controlled on ART (Fig. 4C and D, open symbols with dotted line). In this longitudinal analysis, the CMV-specific CD8 T cells had higher frequencies of cells primed for survival (CC-3lo Bcl-2hi) than the HIV-specific CD8 T cells when the subject was off ART (Fig. 4C). Once HIV viremia was controlled, the frequencies of CMV-specific cells primed for survival or prone to death did not change significantly.
DISCUSSION
The HIV-specific CD8 T cell response is capable of controlling viral replication in acute HIV infection and, rarely, in chronic infection (i.e., long-term nonprogressors [LTNP] and elite controllers) for years. However, this initial response fails to control virus long-term in the majority of chronically infected patients. In the search for correlates of immunity to HIV, several qualities of HIV-specific CD8 T cells have been identified that likely contribute to an effective antiviral immune response. We reason that survival of CD8 T cells may also represent a key factor of an effective immune response. Antigen-presenting cells provide costimulatory signals which are necessary for the generation of polyfunctional CD8 T cell responses and for the maintenance or long-term survival of these responses (44, 45). In the absence of these critical signals, cells are not only less functional with regard to cytokine secretion, proliferation, and lytic capacity but are also more susceptible to apoptosis. Prior studies have demonstrated that HIV-specific CD8 T cells are more prone to apoptosis than CMV-specific CD8 T cells within the same patient (37). The results of the present study demonstrate an important link in HIV immunopathogenesis that may explain in part the inability of HIV-specific CD8 T cells to maintain long-term viral control.
The present study represents the first analysis of cell death and prosurvival factors in a cohort of viremic patients with various levels of viral control, but more importantly, in a cohort of aviremic patients with either immune control (EC) or pharmacologic control (ST) of HIV-1. Similar to previous groups, we demonstrate that HIV-specific CD8 T cells are prone to apoptosis, with viremic patients exhibiting the greatest susceptibility to apoptosis (37). Interestingly, in our cohort, we determined that this was the case regardless of the level of viral control, as we did not observe significant differences in CC-3 levels between viremic progressors with higher viral loads and viremic controllers, those patients capable of maintaining a pVL of <2,000 copies/ml. Despite this high degree of susceptibility to apoptosis, it is notable that chronically infected viremic patients have a high frequency of viral-specific CD8 T cells (46). Some have significant proliferation of viral-specific effector populations, as well as rapid turnover due to apoptosis. Once treated, patients actually have a decline in the frequency of viral-specific CD8 T cells, due to the decrease in antigenic stimulation and proliferation of effector T cells (47).
To our knowledge, this is the first study to also examine aviremic patients, those with VL of <50 copies/ml, who exhibited the lowest levels of CC-3 on HIV-specific CD8 T cells. Interestingly, we found that elite controllers, those patients with immune control of HIV, exhibit lower levels of CC-3 than successfully treated patients with virologic control on ART. These findings suggest that, in chronic infection, the death of HIV-specific CTL may contribute to the loss of viral control and that resistance to apoptosis in CTL from EC may contribute to their ability to intrinsically control viremia. In support of this, longitudinal studies of persons evolving from acute to chronic HIV infection reveal that HIV-specific CD8 T cells with functional attributes associated with viral control, such as high-avidity CTL, are often deleted in chronic infection (48). Similarly, the preservation of immunodominant responses into chronic infection is associated with a lower viral load set point and slower CD4 decline (49).
The susceptibility of CTL to apoptosis is likely related to environmental and intrinsic cellular factors. HIV infection leads to a state of high immune activation, which is created by circulating uncontrolled viremia and nonspecific activation. The pathogenesis and demise of the immune system is attributed in large part to a breakdown in the mucosal barrier and subsequent gut translocation (50). This leads to further cellular activation, replication of virus, and loss of CD4 T cells (51). To better understand how viral and nonviral mechanisms of cell death may contribute to the loss of the CD8 T cell response, we examined the correlation of viral load and expression of CC-3 on CD8 T cells. We attribute one-third of the HIV-specific CD8 T cell death observed to circulating virus (Fig. 5B). The relatively poor correlation with viral load is supported by the observation that progressors with higher viral loads do not exhibit significant differences in expression of CC-3 compared to its expression in viremic controllers, patients capable of viral control to less than 2,000 copies/ml. Furthermore, viral load alone does not explain the observed differences between EC and ST patients, two groups with undetectable circulating plasma virus.
Nonspecific immune activation resulting from gut translocation likely contributes to immune cell death. Indeed, studies of large cohorts of HIV-infected patients have demonstrated a strong correlation of plasma lipopolysaccharide (LPS) with viral load and the activation state of CD8 T cells as measured by HLA-DR and CD38 (50). While gut translocation can account for some of the non-viral-specific causes of cell death in viremic patients, it does not account for the observed differences between EC and ST. If differences in gut translocation and immune activation explain why ST patients have CTL that are more prone to apoptosis, we would expect ST to have higher levels of LPS and immune activation than EC. In fact, this is not the case. In large cohort studies, EC have higher levels of plasma LPS and higher levels of activated CD8 T cells than ST patients (52). Thus, if nonspecific immune activation explained the observed differences in susceptibility, we would expect the EC to have higher frequencies of HIV- and CMV-specific T cell apoptosis than ST. Instead, within any given EC or ST subject, we observed comparable rates of HIV-specific and CMV-specific cell death (Fig. 1D). While beyond the scope of this work, the unique survival advantage of the HIV-specific CD8 T cells in EC may reflect differences in low-level immune activation from viral turnover in cellular reservoirs, as it has been demonstrated that integrated viral DNA is significantly lower in EC than ST (53).
We observed that decreased expression of CD8 on HIV-specific T cells resulted in a greater frequency of cells prone to death, as evidenced by increased expression of CC-3 (Fig. 2). The down-modulation of CD8 expression has been reported in other infection models, including influenza, and can result in T cell nonresponsiveness (54, 55). CD8 down-modulation has been postulated as a mechanism of immune regulation whereby activated T cells become less or nonresponsive. Such a response in a state of persistent viremia like HIV infection likely contributes to disease pathogenesis.
The events preceding cell deletion are an area of intense investigation in the field of chronic viral infections, including HIV infection. While the generation of an effective immune response requires a combination of antigen presentation with costimulatory signals, the persistence of the response depends in part on negative or inhibitory signals. A persistent high antigen load results in progressive CD8 T cell exhaustion, which is characterized by loss of proliferation, ex vivo cytotoxicity, effector cytokine secretion, and eventually, deletion (56–59). Through genome-wide array analysis comparing exhausted CD8 T cells from chronic lymphocytic choriomeningitis virus (LCMV) infection with those generated in acute LCMV infection, molecular signatures have provided insights into the signaling pathways that result in exhaustion and deletion (60). Several inhibitory molecules are overexpressed in exhausted cells, including PD-1, 2B4, TIM-3, Lag-3, and CD160, some of which correlate with clinical parameters of HIV disease progression (60–63). Selective blockade of PD-1 and TIM-3 has been shown to restore effector functions (64, 65). More recent data demonstrate that the accumulation of multiple inhibitory factors, such as double positive CD160 PD-1, is associated with progressive exhaustion and cellular dysfunction (66–68). In contrast, CD8 T cells that express PD-1 without other inhibitory molecules can still recognize cognate antigen and generate a polyfunctional T cell response (68). Similarly, CD8 T cells from EC express mostly single positive CD160 CD8 T cells, suggesting that this phenotype is more functional (68). Future studies incorporating pro- and antiapoptotic parameters with a comprehensive analysis of inhibitory factors would provide insight into the regulation of exhaustion and identify phenotypes which result in irreversible exhaustion and are destined for deletion.
Given the lack of evidence that external factors explain the observed differences in apoptosis between EC and ST, we looked to intrinsic factors to explain how cells might be primed for survival. Costimulatory and cytokine signals (including interleukin-7 [IL-7] and IL-15) regulate T cell homeostasis and the expression of prosurvival molecules in the Bcl-2 family (44, 69–72). In animal models, the timing of costimulatory signals via CD137 monoclonal antibodies (during the first few days of infection with either LCMV or influenzavirus) significantly altered the persistence or deletion of viral-specific CD8 T cells and the subsequent survival of mice (73). This compromised CD8 T cell response was accompanied by higher levels of IL-10 and tumor necrosis factor alpha (TNF-α), which were associated with the loss of CD4 and CD8 T cells, respectively (73). We therefore reason that rapid viral control in acute disease requires an optimal balance of costimulatory and cytokine signals. The observed differences in the survival advantage seen in EC over ST may be a result of the generation of cells primed for survival early in infection. In this scenario, ART-treated patients, despite having undetectable pVL, may have cells prone to apoptosis, in part due to compromised APC and lack of optimal costimulation. To begin to investigate this, we examined the expression of Bcl-2 in memory cells across the cohort. We focused our investigation on memory cells, as effector populations downregulate Bcl-2 expression (43). We found no differences between EC and ST when examining total CD8 T cells, but they had marked differences in HIV-specific CD8 T cells (Fig. 3). In fact, the highest expression was seen in elite controllers, with observed levels twice those seen in the ST cohort. Finally, colabeling analysis of CC-3 and Bcl-2 demonstrated a phenotype of cells primed for survival (CC-3lo Bcl-2hi) (Fig. 4B). In these analyses, EC possessed a larger population of HIV-specific CD8 T cells that are resistant to apoptosis by upregulation of antiapoptotic molecules. In the longitudinal analysis of the viremic patients before and after ART initiation, we demonstrated that although ART can result in upregulation of Bcl-2 on HIV-specific CD8 T cells, the levels in successfully treated patients never achieved the levels seen in EC. This finding suggests that the HIV-specific T cells were generated earlier, prior to viral control, rather than de novo after successful treatment with ART. Finally, to elucidate whether priming for survival was restricted to “good” alleles, such as B57 and B27, we analyzed responses restricted by a favorable HLA, B*57, and by the product of a neutral allele, A*2. We did not observe any selective advantage based on HLA alone. This observation is promising as we translate these data into vaccine development, where an optimal response would be desired in patients of any HLA type.
Our analysis of CMV-specific CD8 T cells demonstrates that (i) CMV-specific T cells from HIV-seropositive subjects are more prone to apoptosis than CMV-specific CD8 T cells from HIV-seronegative subjects (Fig. 1B), (ii) there is a range, albeit modest, of susceptibility to apoptosis of CMV-specific CD8 T cells across the HIV cohort (Fig. 1D), and (iii) ART does not significantly change the frequency of either cells prone to death (CC-3hi Bcl-2lo) or cells primed for survival (CC-3lo Bcl-2hi) (Fig. 4C and D, open symbols). Collectively, these data demonstrate that the survival of CMV-specific CD8 T cells is not compromised to the same degree as that of HIV-specific T cells within the same host. Interestingly, increasing data suggest that CMV-specific T cells in aging and HIV-infected populations are relatively resistant to apoptosis, leading to higher frequencies of CMV-specific T cells in these populations, a phenomenon which may contribute to immunosenescence and inflammation (74).
In summary, the results of the present study demonstrate differences in the survival of CTL in EC, a phenotype that is important for viral control. This work adds to a growing body of knowledge of the generation and maintenance of effective immune responses to HIV. Further work elucidating the pathways for achieving these responses will allow us to gain insight into mechanisms for manipulating the immune response to induce long-lived responses in vaccine recipients.
ACKNOWLEDGMENTS
We thank the volunteers for their participation, Marion Spell for help with flow cytometric acquisition, the Center for AIDS Research repository, and Sarah Seevers and Tiffanie Mann for patient sample processing.
This work was supported by NIH grants NIAID 1 K08 AI076056-01 to S.L.H. and 2 R01 AI064060 and 1 R01 AI084772 to P.A.G.
Footnotes
Published ahead of print 28 February 2013
REFERENCES
- 1. Jin X, Bauer DE, Tuttleton SE, Lewin S, Gettie A, Blanchard J, Irwin CE, Safrit JT, Mittler J, Weinberger L, Kostrikis LG, Zhang L, Perelson AS, Ho DD. 1999. Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 189:991–998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. 1994. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 68:4650–4655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Carrington M, Nelson GW, Martin MP, Kissner T, Vlahov D, Goedert JJ, Kaslow R, Buchbinder S, Hoots K, O'Brien SJ. 1999. HLA and HIV-1: heterozygote advantage and B*35-Cw*04 disadvantage. Science 283:1748–1752 [DOI] [PubMed] [Google Scholar]
- 4. Hendel H, Caillat-Zucman S, Lebuanec H, Carrington M, O'Brien S, Andrieu JM, Schachter F, Zagury D, Rappaport J, Winkler C, Nelson GW, Zagury JF. 1999. New class I and II HLA alleles strongly associated with opposite patterns of progression to AIDS. J. Immunol. 162:6942–6946 [PubMed] [Google Scholar]
- 5. Kaslow RA, Carrington M, Apple R, Park L, Munoz A, Saah AJ, Goedert JJ, Winkler C, O'Brien SJ, Rinaldo C, Detels R, Blattner W, Phair J, Erlich H, Mann DL. 1996. Influence of combinations of human major histocompatibility complex genes on the course of HIV-1 infection. Nat. Med. 2:405–411 [DOI] [PubMed] [Google Scholar]
- 6. MacDonald KS, Fowke KR, Kimani J, Dunand VA, Nagelkerke NJ, Ball TB, Oyugi J, Njagi E, Gaur LK, Brunham RC, Wade J, Luscher MA, Krausa P, Rowland-Jones S, Ngugi E, Bwayo JJ, Plummer FA. 2000. Influence of HLA supertypes on susceptibility and resistance to human immunodeficiency virus type 1 infection. J. Infect. Dis. 181:1581–1589 [DOI] [PubMed] [Google Scholar]
- 7. McNeil AJ, Yap PL, Gore SM, Brettle RP, McColl M, Wyld R, Davidson S, Weightman R, Richardson AM, Robertson JR. 1996. Association of HLA types A1-B8-DR3 and B27 with rapid and slow progression of HIV disease. QJM 89:177–185 [DOI] [PubMed] [Google Scholar]
- 8. Addo MM, Yu XG, Rathod A, Cohen D, Eldridge RL, Strick D, Johnston MN, Corcoran C, Wurcel AG, Fitzpatrick CA, Feeney ME, Rodriguez WR, Basgoz N, Draenert R, Stone DR, Brander C, Goulder PJ, Rosenberg ES, Altfeld M, Walker BD. 2003. 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. 77:2081–2092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bansal A, Sabbaj S, Edwards BH, Ritter D, Perkins C, Tang J, Szinger JJ, Weiss H, Goepfert PA, Korber B, Wilson CM, Kaslow RA, Mulligan MJ. 2003. T cell responses in HIV type 1-infected adolescent minorities share similar epitope specificities with whites despite significant differences in HLA class I alleles. AIDS Res. Hum. Retroviruses 19:1017–1026 [DOI] [PubMed] [Google Scholar]
- 10. Edwards BH, Bansal A, Sabbaj S, Bakari J, Mulligan MJ, Goepfert PA. 2002. Magnitude of functional CD8+ T-cell responses to the gag protein of human immunodeficiency virus type 1 correlates inversely with viral load in plasma. J. Virol. 76:2298–2305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ogg GS, Jin X, Bonhoeffer S, Dunbar PR, Nowak MA, Monard S, Segal JP, Cao Y, Rowland-Jones SL, Cerundolo V, Hurley A, Markowitz M, Ho DD, Nixon DF, McMichael AJ. 1998. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 279:2103–2106 [DOI] [PubMed] [Google Scholar]
- 12. Almeida JR, Price DA, Papagno L, Arkoub ZA, Sauce D, Bornstein E, Asher TE, Samri A, Schnuriger A, Theodorou I, Costagliola D, Rouzioux C, Agut H, Marcelin AG, Douek D, Autran B, Appay V. 2007. Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. J. Exp. Med. 204:2473–2485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA, Abraham J, Lederman MM, Benito JM, Goepfert PA, Connors M, Roederer M, Koup RA. 2006. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood 107:4781–4789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Critchfield JW, Young DH, Hayes TL, Braun JV, Garcia JC, Pollard RB, Shacklett BL. 2008. Magnitude and complexity of rectal mucosa HIV-1-specific CD8+ T-cell responses during chronic infection reflect clinical status. PLoS One 3:e3577 doi:10.1371/journal.pone.0003577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Horton H, Frank I, Baydo R, Jalbert E, Penn J, Wilson S, McNevin JP, McSweyn MD, Lee D, Huang Y, De Rosa SC, McElrath MJ. 2006. Preservation of T cell proliferation restricted by protective HLA alleles is critical for immune control of HIV-1 infection. J. Immunol. 177:7406–7415 [DOI] [PubMed] [Google Scholar]
- 16. Migueles SA, Laborico AC, Shupert WL, Sabbaghian MS, Rabin R, Hallahan CW, Van Baarle D, Kostense S, Miedema F, McLaughlin M, Ehler L, Metcalf J, Liu S, Connors M. 2002. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat. Immunol. 3:1061–1068 [DOI] [PubMed] [Google Scholar]
- 17. Appay V, Nixon DF, Donahoe SM, Gillespie GM, Dong T, King A, Ogg GS, Spiegel HM, Conlon C, Spina CA, Havlir DV, Richman DD, Waters A, Easterbrook P, McMichael AJ, Rowland-Jones SL. 2000. HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J. Exp. Med. 192:63–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Benito JM, Lopez M, Soriano V. 2004. The role of CD8+ T-cell response in HIV infection. AIDS Rev. 6:79–88 [PubMed] [Google Scholar]
- 19. Betts MR, Ambrozak DR, Douek DC, Bonhoeffer S, Brenchley JM, Casazza JP, Koup RA, Picker LJ. 2001. Analysis of total human immunodeficiency virus (HIV)-specific CD4(+) and CD8(+) T-cell responses: relationship to viral load in untreated HIV infection. J. Virol. 75:11983–11991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Day CL, Kiepiela P, Leslie AJ, van der Stok M, Nair K, Ismail N, Honeyborne I, Crawford H, Coovadia HM, Goulder PJ, Walker BD, Klenerman P. 2007. Proliferative capacity of epitope-specific CD8 T-cell responses is inversely related to viral load in chronic human immunodeficiency virus type 1 infection. J. Virol. 81:434–438 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Jagannathan P, Osborne CM, Royce C, Manion MM, Tilton JC, Li L, Fischer S, Hallahan CW, Metcalf JA, McLaughlin M, Pipeling M, McDyer JF, Manley TJ, Meier JL, Altman JD, Hertel L, Davey RT, Jr, Connors M, Migueles SA. 2009. Comparisons of CD8+ T cells specific for human immunodeficiency virus, hepatitis C virus, and cytomegalovirus reveal differences in frequency, immunodominance, phenotype, and interleukin-2 responsiveness. J. Virol. 83:2728–2742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Goulder PJ, Phillips RE, Colbert RA, McAdam S, Ogg G, Nowak MA, Giangrande P, Luzzi G, Morgan B, Edwards A, McMichael AJ, Rowland-Jones S. 1997. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nat. Med. 3:212–217 [DOI] [PubMed] [Google Scholar]
- 23. Katsikis PD, Wunderlich ES, Smith CA, Herzenberg LA, Herzenberg LA. 1995. Fas antigen stimulation induces marked apoptosis of T lymphocytes in human immunodeficiency virus-infected individuals. J. Exp. Med. 181:2029–2036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Petrovas C, Chaon B, Ambrozak DR, Price DA, Melenhorst JJ, Hill BJ, Geldmacher C, Casazza JP, Chattopadhyay PK, Roederer M, Douek DC, Mueller YM, Jacobson JM, Kulkarni V, Felber BK, Pavlakis GN, Katsikis PD, Koup RA. 2009. Differential association of programmed death-1 and CD57 with ex vivo survival of CD8+ T cells in HIV infection. J. Immunol. 183:1120–1132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Petrovas C, Mueller YM, Katsikis PD. 2005. Apoptosis of HIV-specific CD8+ T cells: an HIV evasion strategy. Cell Death Differ. 12(Suppl 1):859–870 [DOI] [PubMed] [Google Scholar]
- 26. Yamamoto T, Price DA, Casazza JP, Ferrari G, Nason M, Chattopadhyay PK, Roederer M, Gostick E, Katsikis PD, Douek DC, Haubrich R, Petrovas C, Koup RA. 2011. Surface expression patterns of negative regulatory molecules identify determinants of virus-specific CD8+ T-cell exhaustion in HIV infection. Blood 117:4805–4815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, de Souza M, Adams E, Benenson M, Gurunathan S, Tartaglia J, McNeil JG, Francis DP, Stablein D, Birx DL, Chunsuttiwat S, Khamboonruang C, Thongcharoen P, Robb ML, Michael NL, Kunasol P, Kim JH. 2009. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 361:2209–2220 [DOI] [PubMed] [Google Scholar]
- 28. Putcha GV, Harris CA, Moulder KL, Easton RM, Thompson CB, Johnson EM., Jr 2002. Intrinsic and extrinsic pathway signaling during neuronal apoptosis: lessons from the analysis of mutant mice. J. Cell Biol. 157:441–453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Danial NN, Korsmeyer SJ. 2004. Cell death: critical control points. Cell 116:205–219 [DOI] [PubMed] [Google Scholar]
- 30. Opferman JT, Korsmeyer SJ. 2003. Apoptosis in the development and maintenance of the immune system. Nat. Immunol. 4:410–415 [DOI] [PubMed] [Google Scholar]
- 31. Shacka JJ, Roth KA. 2006. Bcl-2 family and the central nervous system: from rheostat to real complex. Cell Death Differ. 13:1299–1304 [DOI] [PubMed] [Google Scholar]
- 32. Salvesen GS, Dixit VM. 1997. Caspases: intracellular signaling by proteolysis. Cell 91:443–446 [DOI] [PubMed] [Google Scholar]
- 33. Gougeon ML, Lecoeur H, Dulioust A, Enouf MG, Crouvoiser M, Goujard C, Debord T, Montagnier L. 1996. Programmed cell death in peripheral lymphocytes from HIV-infected persons: increased susceptibility to apoptosis of CD4 and CD8 T cells correlates with lymphocyte activation and with disease progression. J. Immunol. 156:3509–3520 [PubMed] [Google Scholar]
- 34. Zaunders JJ, Moutouh-de Parseval L, Kitada S, Reed JC, Rought S, Genini D, Leoni L, Kelleher A, Cooper DA, Smith DE, Grey P, Estaquier J, Little S, Richman DD, Corbeil J. 2003. Polyclonal proliferation and apoptosis of CCR5+ T lymphocytes during primary human immunodeficiency virus type 1 infection: regulation by interleukin (IL)-2, IL-15, and Bcl-2. J. Infect. Dis. 187:1735–1747 [DOI] [PubMed] [Google Scholar]
- 35. Liegler TJ, Yonemoto W, Elbeik T, Vittinghoff E, Buchbinder SP, Greene WC. 1998. Diminished spontaneous apoptosis in lymphocytes from human immunodeficiency virus-infected long-term nonprogressors. J. Infect. Dis. 178:669–679 [DOI] [PubMed] [Google Scholar]
- 36. Moretti S, Marcellini S, Boschini A, Famularo G, Santini G, Alesse E, Steinberg SM, Cifone MG, Kroemer G, De Simone C. 2000. Apoptosis and apoptosis-associated perturbations of peripheral blood lymphocytes during HIV infection: comparison between AIDS patients and asymptomatic long-term non-progressors. Clin. Exp. Immunol. 122:364–373 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Mueller YM, De Rosa SC, Hutton JA, Witek J, Roederer M, Altman JD, Katsikis PD. 2001. Increased CD95/Fas-induced apoptosis of HIV-specific CD8(+) T cells. Immunity 15:871–882 [DOI] [PubMed] [Google Scholar]
- 38. Petrovas C, Mueller YM, Dimitriou ID, Bojczuk PM, Mounzer KC, Witek J, Altman JD, Katsikis PD. 2004. HIV-specific CD8+ T cells exhibit markedly reduced levels of Bcl-2 and Bcl-xL. J. Immunol. 172:4444–4453 [DOI] [PubMed] [Google Scholar]
- 39. Bansal A, Gough E, Sabbaj S, Ritter D, Yusim K, Sfakianos G, Aldrovandi G, Kaslow RA, Wilson CM, Mulligan MJ, Kilby JM, Goepfert PA. 2005. CD8 T-cell responses in early HIV-1 infection are skewed towards high entropy peptides. AIDS 19:241–250 [PubMed] [Google Scholar]
- 40. Bansal A, Yue L, Conway J, Yusim K, Tang J, Kappes J, Kaslow RA, Wilson CM, Goepfert PA. 2007. Immunological control of chronic HIV-1 infection: HLA-mediated immune function and viral evolution in adolescents. AIDS 21:2387–2397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Fuller MJ, Hildeman DA, Sabbaj S, Gaddis DE, Tebo AE, Shang L, Goepfert PA, Zajac AJ. 2005. Cutting edge: emergence of CD127high functionally competent memory T cells is compromised by high viral loads and inadequate T cell help. J. Immunol. 174:5926–5930 [DOI] [PubMed] [Google Scholar]
- 42. Sabbaj S, Heath SL, Bansal A, Vohra S, Kilby JM, Zajac AJ, Goepfert PA. 2007. Functionally competent antigen-specific CD127(hi) memory CD8+ T cells are preserved only in HIV-infected individuals receiving early treatment. J. Infect. Dis. 195:108–117 [DOI] [PubMed] [Google Scholar]
- 43. Grayson JM, Zajac AJ, Altman JD, Ahmed R. 2000. Cutting edge: increased expression of Bcl-2 in antigen-specific memory CD8+ T cells. J. Immunol. 164:3950–3954 [DOI] [PubMed] [Google Scholar]
- 44. Vinay DS, Kwon BS. 1998. Role of 4-1BB in immune responses. Semin. Immunol. 10:481–489 [DOI] [PubMed] [Google Scholar]
- 45. Wen T, Bukczynski J, Watts TH. 2002. 4-1BB ligand-mediated costimulation of human T cells induces CD4 and CD8 T cell expansion, cytokine production, and the development of cytolytic effector function. J. Immunol. 168:4897–4906 [DOI] [PubMed] [Google Scholar]
- 46. Draenert R, Verrill CL, Tang Y, Allen TM, Wurcel AG, Boczanowski M, Lechner A, Kim AY, Suscovich T, Brown NV, Addo MM, Walker BD. 2004. Persistent recognition of autologous virus by high-avidity CD8 T cells in chronic, progressive human immunodeficiency virus type 1 infection. J. Virol. 78:630–641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Casazza JP, Betts MR, Picker LJ, Koup RA. 2001. Decay kinetics of human immunodeficiency virus-specific CD8+ T cells in peripheral blood after initiation of highly active antiretroviral therapy. J. Virol. 75:6508–6516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Lichterfeld M, Yu XG, Mui SK, Williams KL, Trocha A, Brockman MA, Allgaier RL, Waring MT, Koibuchi T, Johnston MN, Cohen D, Allen TM, Rosenberg ES, Walker BD, Altfeld M. 2007. Selective depletion of high-avidity human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T cells after early HIV-1 infection. J. Virol. 81:4199–4214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Streeck H, Jolin JS, Qi Y, Yassine-Diab B, Johnson RC, Kwon DS, Addo MM, Brumme C, Routy JP, Little S, Jessen HK, Kelleher AD, Hecht FM, Sekaly RP, Rosenberg ES, Walker BD, Carrington M, Altfeld M. 2009. 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. 83:7641–7648 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, Blazar BR, Rodriguez B, Teixeira-Johnson L, Landay A, Martin JN, Hecht FM, Picker LJ, Lederman MM, Deeks SG, Douek DC. 2006. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 12:1365–1371 [DOI] [PubMed] [Google Scholar]
- 51. Brenchley JM, Douek DC. 2008. The mucosal barrier and immune activation in HIV pathogenesis. Curr. Opin. HIV AIDS 3:356–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, Page-Shafer K, Hsue P, Emu B, Krone M, Lampiris H, Douek D, Martin JN, Deeks SG. 2008. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J. Infect. Dis. 197:126–133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Graf EH, Mexas AM, Yu JJ, Shaheen F, Liszewski MK, Di Mascio M, Migueles SA, Connors M, O'Doherty U. 2011. Elite suppressors harbor low levels of integrated HIV DNA and high levels of 2-LTR circular HIV DNA compared to HIV+ patients on and off HAART. PLoS Pathog. 7:e1001300 doi:10.1371/journal.ppat.1001300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Robbins PA, McMichael AJ. 1991. Immune recognition of HLA molecules downmodulates CD8 expression on cytotoxic T lymphocytes. J. Exp. Med. 173:221–230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Takada K, Jameson SC. 2009. Self-class I MHC molecules support survival of naive CD8 T cells, but depress their functional sensitivity through regulation of CD8 expression levels. J. Exp. Med. 206:2253–2269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Fuller MJ, Zajac AJ. 2003. Ablation of CD8 and CD4 T cell responses by high viral loads. J. Immunol. 170:477–486 [DOI] [PubMed] [Google Scholar]
- 57. van der Most RG, Murali-Krishna K, Lanier JG, Wherry EJ, Puglielli MT, Blattman JN, Sette A, Ahmed R. 2003. Changing immunodominance patterns in antiviral CD8 T-cell responses after loss of epitope presentation or chronic antigenic stimulation. Virology 315:93–102 [DOI] [PubMed] [Google Scholar]
- 58. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. 2003. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77:4911–4927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, Ahmed R. 1998. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188:2205–2213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Wherry EJ, Ha SJ, Kaech SM, Haining WN, Sarkar S, Kalia V, Subramaniam S, Blattman JN, Barber DL, Ahmed R. 2007. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27:670–684 [DOI] [PubMed] [Google Scholar]
- 61. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. 2006. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443:350–354 [DOI] [PubMed] [Google Scholar]
- 62. Jones RB, Ndhlovu LC, Barbour JD, Sheth PM, Jha AR, Long BR, Wong JC, Satkunarajah M, Schweneker M, Chapman JM, Gyenes G, Vali B, Hyrcza MD, Yue FY, Kovacs C, Sassi A, Loutfy M, Halpenny R, Persad D, Spotts G, Hecht FM, Chun TW, McCune JM, Kaul R, Rini JM, Nixon DF, Ostrowski MA. 2008. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 205:2763–2779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, Precopio ML, Schacker T, Roederer M, Douek DC, Koup RA. 2006. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J. Exp. Med. 203:2281–2292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, Castelblanco N, Kuchroo V, Gretch DR, Rosen HR. 2009. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J. Virol. 83:9122–9130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, Routy JP, Haddad EK, Sekaly RP. 2006. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 12:1198–1202 [DOI] [PubMed] [Google Scholar]
- 66. Bengsch B, Seigel B, Ruhl M, Timm J, Kuntz M, Blum HE, Pircher H, Thimme R. 2010. Coexpression of PD-1, 2B4, CD160 and KLRG1 on exhausted HCV-specific CD8+ T cells is linked to antigen recognition and T cell differentiation. PLoS Pathog. 6:e1000947 doi:10.1371/journal.ppat.1000947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. 2009. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10:29–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Peretz Y, He Z, Shi Y, Yassine-Diab B, Goulet JP, Bordi R, Filali-Mouhim A, Loubert JB, El-Far M, Dupuy FP, Boulassel MR, Tremblay C, Routy JP, Bernard N, Balderas R, Haddad EK, Sekaly RP. 2012. CD160 and PD-1 co-expression on HIV-specific CD8 T cells defines a subset with advanced dysfunction. PLoS Pathog. 8:e1002840 doi:10.1371/journal.ppat.1002840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Boise LH, Minn AJ, Noel PJ, June CH, Accavitti MA, Lindsten T, Thompson CB. 1995. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity 3:87–98 [DOI] [PubMed] [Google Scholar]
- 70. Bukczynski J, Wen T, Watts TH. 2003. Costimulation of human CD28-T cells by 4-1BB ligand. Eur. J. Immunol. 33:446–454 [DOI] [PubMed] [Google Scholar]
- 71. Lee HW, Park SJ, Choi BK, Kim HH, Nam KO, Kwon BS. 2002. 4-1BB promotes the survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. J. Immunol. 169:4882–4888 [DOI] [PubMed] [Google Scholar]
- 72. Starck L, Scholz C, Dorken B, Daniel PT. 2005. Costimulation by CD137/4-1BB inhibits T cell apoptosis and induces Bcl-xL and c-FLIP(short) via phosphatidylinositol 3-kinase and AKT/protein kinase B. Eur. J. Immunol. 35:1257–1266 [DOI] [PubMed] [Google Scholar]
- 73. Zhang B, Maris CH, Foell J, Whitmire J, Niu L, Song J, Kwon BS, Vella AT, Ahmed R, Jacob J, Mittler RS. 2007. Immune suppression or enhancement by CD137 T cell costimulation during acute viral infection is time dependent. J. Clin. Invest. 117:3029–3041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Naeger DM, Martin JN, Sinclair E, Hunt PW, Bangsberg DR, Hecht F, Hsue P, McCune JM, Deeks SG. 2010. Cytomegalovirus-specific T cells persist at very high levels during long-term antiretroviral treatment of HIV disease. PLoS One 5:e8886 doi:10.1371/journal.pone.0008886 [DOI] [PMC free article] [PubMed] [Google Scholar]





