Abstract
Human immunodeficiency virus (HIV)-1 causes T cell anergy and affects T cell maturation. Various mechanisms are responsible for impaired anti-HIV-1-specific responses: programmed death (PD)-1 molecule and its ligand PD-L1 are negative regulators of T cell activity and their expression is increased during HIV-1 infection. This study examines correlations between T cell maturation, expression of PD-1 and PD-L1, and the effects of their blockade. Peripheral blood mononuclear cells (PBMC) from 24 HIV-1+ and 17 uninfected individuals were phenotyped for PD-1 and PD-L1 expression on CD4+ and CD8+ T cell subsets. The effect of PD-1 and PD-L1 blockade on proliferation and interferon (IFN)-γ production was tested on eight HIV-1+ patients. Naive (CCR7+CD45RA+) CD8+ T cells were reduced in HIV-1 aviraemic (P = 0·0065) and viraemic patients (P = 0·0130); CD8 T effector memory subsets [CCR7−CD45RA–(TEM)] were increased in HIV-1+ aviraemic (P = 0·0122) and viraemic (P = 0·0023) individuals versus controls. PD-1 expression was increased in CD4 naive (P = 0·0496), central memory [CCR7+CD45RA– (TCM); P = 0·0116], TEM (P = 0·0037) and CD8 naive T cells (P = 0·0133) of aviraemic HIV-1+versus controls. PD-L1 was increased in CD4 TEMRA (CCR7−CD45RA+, P = 0·0119), CD8 TEM (P = 0·0494) and CD8 TEMRA (P = 0·0282) of aviraemic HIV-1+versus controls. PD-1 blockade increased HIV-1-specific proliferative responses in one of eight patients, whereas PD-L1 blockade restored responses in four of eight patients, but did not increase IFN-γ-production. Alteration of T cell subsets, accompanied by increased PD-1 and PD-L1 expression in HIV-1 infection contributes to anergy and impaired anti-HIV-1-specific responses which are not rescued when PD-1 is blocked, in contrast to when PD-L1 is blocked, due possibly to an ability to bind to receptors other than PD-1.
Keywords: anergy, HIV-1, PD-1, PD-L1, T cell subsets
Introduction
T cell anergy and clonal exhaustion are characteristic defects of chronic human immunodeficiency virus (HIV)-1 infection and account for the inability of the immune system to mount an effective response to the virus and ultimately clear it from the organism. Reduced interferon (IFN)-γ production and low proliferative capacity in response to stimulus are signs of this defect, even in patients receiving highly active anti-retroviral therapy (HAART) [1,2]. Various mechanisms are responsible for the defective anti-HIV-1 immune response. Ligation of the T cell receptor (TCR) complex is crucial for T cell activation, and is regulated by co-stimulatory molecules which can provide positive and negative signalling. Positive signals are mediated by molecules such as CD28, which is expressed constitutively on T cells and upon ligation is able to induce T cell proliferation and cytokine production [3,4].
The programmed death 1 (PD-1) molecule is an inducible negative regulator of T cell activity [5]. PD-1 is expressed on lymphocytes and in particular on T and B cells. PD-L1 is one of the ligands for PD-1 and is expressed both on antigen- presenting cells (APC) and T cells. Recent in vitro and in vivo studies, including a mouse model of choriomeningitis, have shown the importance of the PD-1/PD-L1 interaction in chronic viral infection [6–8]. In HIV-1+ patients, levels of PD-1 and PD-L1 are increased even when patients are receiving successful HAART and have undetectable viral loads, suggesting that the increase in the expression of these two molecules is linked to HIV-1 related anergy [7,9,10].
CD4+ T cells are the main target for HIV-1 infection [11]; their number is reduced progressively and their function impaired during the course of disease [12]. Differentiation/maturation of CD8+cytotoxic T lymphocytes (CTL), which are crucial for anti-viral action, is also affected by HIV-1 infection due in part to lack of help from CD4+ T cells [13]. The differentiation stages of CD4+ and CD8+ T cells can be categorized phenotypically according to the expression of CD45RA and CCR7. Four different subsets of T cells have been identified [13–15]: naive T cells (CCR7+CD45RA+); central memory T cells (TCM) (CCR7+, CD45RA–); effector memory T cells (TEM) (CCR7−CD45RA–); and terminally differentiated effector T cells (TEMRA) (CCR7−CD45RA+). Naive T cells following priming can differentiate first into effector/effector memory cells and subsequently become TCM. TCM are cells able to proliferate and produce cytokines such as interleukin (IL)-2, and because of their ability to replicate and differentiate they are crucial for the maintenance of the memory pool. TCM are affected particularly during HIV-1 infection [15], especially during the acute stage of disease during which they are rapidly depleted [12], but they can be restored partially by HAART [1,2]. TEM are less able to divide and produce IL-2 but are characterized by the ability to produce IFN-γ and perforin during viral infection. TEMRA are maturated further than the TEM phenotype and are the T cells which are least prone to divide. TEM and TEMRA subsets' relative percentages are increased during HIV-1 infection, especially within the HIV-1-specific CD8+ T cell compartment [13].
In this study we investigated expression of PD-1 and PD-L1 on different CD4+ and CD8+ T cell subsets in HIV-1+ patients and healthy controls to determine whether PD-1 and PD-L1 expression may correlate with the abnormal distribution of subsets of CD4+ and CD8+ T cells, and whether expression may be related to HIV-1-induced anergy. We also attempted blockade of PD-1 and PD-L1 to assess whether HIV-1-specific T cell responses would be rescued in vitro.
Materials and methods
Study cohort
Blood samples taken from 17 healthy volunteers and 24 HIV-1+patients were used in the phenotypical study. Eighteen patients were on HAART and had plasma HIV-1 RNA below the detection limit (50 copies/ml), with median CD4 T cell counts of 412 cells/µl blood [interquartile range (IQR) 225–771]. The remaining six patients were treated with HAART but had detectable viral loads (median 16826 IQR 97–68831). The median CD4 count for this second group was 231 cells/µl blood (IQR 37–248). For the functional studies, consisting of lymphocyte proliferation assays (LPR) and enzyme-linked immunospot (ELISPOT) assays, peripheral blood mononuclear cells (PBMC) from eight HAART-treated HIV-1+ patients were used. All but two patients had undetectable viral load. Detailed patients' characteristics for each group are shown in Table 1. The patients' informed consent and Ethics Committee approval were obtained for the studies described.
Table 1.
Patients' characteristics.
Phenotyping | CD4 (cells/µl blood) | CD8 (cells/µl blood) | VL (copies/ml) |
---|---|---|---|
Aviraemic | |||
1 | 1436 | 5060 | < 50 |
2 | 97 | 416 | < 50 |
3 | 403 | 832 | < 50 |
4 | 811 | 452 | < 50 |
5 | 1489 | 680 | < 50 |
6 | 1117 | 990 | < 50 |
7 | 120 | 997 | < 50 |
8 | 565 | 391 | < 50 |
9 | 420 | 741 | < 50 |
10 | 157 | 986 | < 50 |
11 | 643 | 1493 | < 50 |
12 | 400 | 680 | < 50 |
13 | 217 | 638 | < 50 |
14 | 758 | 823 | < 50 |
15 | 449 | 1011 | < 50 |
16 | 339 | 745 | < 50 |
17 | 328 | 668 | < 50 |
18 | 228 | 792 | < 50 |
Median (IQR) | 412 (225–771) | 768 (661–992) | |
Viraemic | |||
1 | 255 | 2014 | 37019 |
2 | 246 | 676 | 108 |
3 | 224 | 524 | 31667 |
4 | 8 | 286 | 164267 |
5 | 238 | 691 | 1984 |
6 | 47 | 457 | 64 |
Median (IQR) | 231 (37–248) | 600 (414–1022) | 16826 (97–68831) |
Functional studies | |||
1† | 291 | 1095 | 333 |
2† | 889 | 1221 | < 50 |
3† | 272 | 953 | 1063 |
4† | 763 | 1377 | < 50 |
5† | 97 | 416 | < 50 |
6 | 1489 | 680 | < 50 |
7 | 157 | 986 | < 50 |
8 | 339 | 745 | < 50 |
Median (IQR) | 315 (186–858) | 970 (696–1190) |
Enzyme-linked immunospot assay and lymphocyte proliferative response. IQR: interquartile range; VL: viral load.
Collection and separation of blood
Twenty millilitres of whole blood were collected into lithium heparin tubes (Becton Dickinson, Oxford, UK). Fresh PBMC were separated by density gradient centrifugation using Histopaque-1077 (Sigma, Poole, UK).
Viral load
Plasma viral load was measured using Versant HIV-1 RNA 3·0 branched assay (Siemens Healthcare, Camberley, UK) with a lower detection limit of 50 HIV-1 RNA copies/ml.
Lymphocyte subsets
Whole blood lymphocyte measurement was performed by staining with murine anti-human monoclonal antibodies (mAb) against CD3, CD4 and CD8 (Tetra One, Beckman Coulter, High Wycombe, UK). Analysis was carried out using an FC-500 Beckman Coulter flow cytometer.
PBMC phenotypic analysis
Freshly isolated PBMC (106) were stained for 30 min with murine anti-human mAb: fluorescein isothiocyanate (FITC) PD-1, phycoerythrin (PE)-Cy7 PD-L1, APC CD45RA, peridinin chlorophyll (PerCP) CD3, APC-Cy7 CD8 (Becton Dickinson) and PE CCR7 (R&D Systems, Abingdon, UK). Matched isotype controls were used for each fluorochrome analysed. Multi-colour flow cytometric analysis was performed on an LSRII six-colour flow cytometer (Becton Dickinson) using Diva software (Becton Dickinson) for acquisition and post-acquisition analysis.
Antigens and peptide pools
Peptide pools used for the proliferation assays and IFN-γ ELISPOT assays were a pool of HIV-1 Gag p24 20mer overlapping peptides (NIBSC, Potters Bar, UK), and a pool of 33 HIV-1 major histocompatibility complex (MHC) class I restricted Gag p17 and p24 9mer peptides (Sigma-Genosys Ltd, Cambridge, UK). The peptide pools were used at a final concentration of 5 µg/ml. Phytohaemoagglutinin (PHA) (Sigma) was used as a positive control at a concentration of 5 µg/ml [16,17].
Blocking antibodies
Murine anti-human PD-1 (clone J116) and PD-L1 (clone MIH1) and matched isotype control antibodies (eBiosciences, San Diego, CA, USA) were used for the blocking experiments at a final concentration of 10 µg/ml.
Proliferation assay
Freshly isolated PBMC (105) were cultured for 5 days in 10% AB plasma/RPMI-1640 (Sigma), with either 5 µg/ml Gag 20mer pool, 5 µg/ml Gag 9mer pool, 5 µg/ml PHA (positive control) or 10% AB plasma/RPMI-1640 only (negative control), in round-bottomed microtitre plates (Greiner, Stonehouse, UK). Cultures were performed in the presence of 10 µg/ml of PD-1 or PD-L1 blocking antibody, isotype control or phosphate-buffered saline (PBS), the latter used as negative control (no difference was found between isotype control antibody or PBS; data not shown). After 5 days each well was pulsed with 1 µCi [3H]-methylthymidine [3H]-TdR (Amersham International, Amersham, UK) and after a further 6 h incubation cells were harvested onto glass fibre filtermats (Wallac Oy, Turku, Finland). Proliferation was measured by liquid scintillation spectroscopy using a 1205 Betaplate counter (Wallac Oy). A positive response is defined as a stimulation index score (obtained by dividing the experimental value by the value of the tissue culture media negative control) of ≥ 3 coupled with a Δ count per minute of > 600.
IFN-γ ELISPOT assays
Detection of single cell release of IFN-γ in ELISPOT assays was carried out as recommended by the manufacturer (Mabtech, Nacka Strand, Sweden) and as described previously [16]. All assays were performed in duplicate with freshly isolated PBMC. Polyvinylidene difluoride (PVDF) 96-well plates (Millipore, Livingston, UK) were coated with 100 µl anti-IFN-γ mAb at 10 µg/ml (Mabtech) overnight at 4°C. Cells were stimulated with Gag 20mer, Gag 9mer peptide pools or 10% AB plasma/RPMI-1640 in the presence of 10 µg/ml of PD-1 or PD-L1 blocking antibody, isotype control antibody or PBS (negative controls). Plates were incubated for 20 h at 37°C in 5% CO2. After the incubation plates were washed, 100 µl biotinylated anti-IFN-γ (Mabtech) was added at 1 µg/ml in PBS to each well, and plates incubated for 2 h at room temperature. After washing, 100 µl streptavidin–alkaline phosphatase (ALP) antibody (Mabtech) was added at 1/1000 dilution in PBS to each well for a final incubation at room temperature for 1 h. Plates were washed, developed and counted. Background negative control values were < 30 spot-forming cells (SFC) per million PBMC. PHA was used as a positive control and values of > 250 SFC/million PBMC were obtained in all assays.
Statistical analysis
Lymphocyte subsets, viral loads and immune response data were not distributed normally. GraphPad5 (GraphPad Software, La Jolla, CA, USA) was used for all statistical calculations. Initial analysis of data was performed using the t-test and further analysis was carried out using the Mann–Whitney U-test. P-values below 0·05 were considered significant.
Results
Distribution of T cell memory subsets
First we investigated the distribution of CD4+ and CD8+ T cells among the different memory subsets according to the expression of CCR7 and CD45RA (an example of the gating strategy is shown in Fig. 1a).
Fig. 1.
The plots show gating strategy for T cell memory subsets on CD4 and CD8 T cells of a representative human immunodeficiency virus (HIV)-1+ aviraemic patient (a). The graphs show combined data for flow cytometric analysis of distribution of naive, central memory T cells (TCM), effector memory T cells (TEM) and terminally differentiated effector T cells (TEMRA) subsets in CD4+ (b) and CD8+ (c) T cells in healthy donors (▭) compared to HIV-1+ aviraemic patients () and HIV-1+ viraemic patients (
). Box plots on graphs show median (central bar), interquartile range (IQR) (box), 10th and 90th percentile (whiskers) and outliers (black dots).
We observed no significant difference in the distribution in any of the CD4+ T cell subsets. However, we found a trend to increased percentages of TEM in both the viraemic and aviraemic HIV-1+ groups compared to healthy controls. We also observed a slight reduction in the naive CD4+ T cell subsets in HIV-1+ patients when compared with data from healthy donors (Fig. 1b). When the CD8+ T cell compartment was analysed we found a reduction in the frequency of cells in the naive T cell compartment in aviraemic HIV-1+ (P = 0·0065) and viraemic HIV-1+ patients (P = 0·0130) compared with healthy controls. We also found an increased percentage of TEM in the viraemic HIV-1+ (P = 0·0023) and in the aviraemic HIV-1+ (P = 0·0122) compared to controls (Fig. 1c).
Levels of PD-1 expression on CD4 and CD8 T cell subsets
We measured the expression of PD-1 in the different CD4+ and CD8+ T cell subsets (examples of PD-1 expression profiles are shown in Fig. 2f). We found an increase in the percentage of CD4+ cells expressing PD-1 in the TCM subset in aviraemic HIV-1+ patients compared to healthy controls (P = 0·0116) (Fig. 2a). PD-1-expressing TCM cells were also increased in the viraemic group, but the difference did not reach statistical significance. Expression of PD-1 was also increased in the CD4+ subset of TEM in aviraemic patients compared to healthy controls (P = 0·0037), and in the CD4+naive subset in aviraemic patients (P = 0·0496) compared to controls (Fig. 2a). No differences were observed between the groups when CD4+ TEMRA were analysed. Mean fluorescence intensity (MFI) for PD-1 did not differ between the three different groups, although there was a trend towards increased PD-1 expression in both aviraemic and viraemic patients in different CD4 subsets (Fig. 2b). In the CD8+T cell subsets we found a statistically significant increase in the percentages of naive T cells expressing PD-1 in the aviraemic patient groups (P = 0·0133) (Fig. 2c). Again, MFI for PD-1 did not differ between the three different groups of CD8 T cell subsets (Fig. 2d).
Fig. 2.
Cumulative data for flow cytometric analysis of expression of programmed death (PD)-1 molecule on the different memory subsets on CD4+[a: percentages; b: mean fluorescence intensity (MFI)] and CD8+ T cells (c: percentages; d: MFI). Bar graphs show the mean, and error bars show standard error of the mean, in the three different groups: healthy controls (▭), aviraemic patients () and viraemic patients (
). Histograms show PD-1 expression on CD4 and CD8 T cells from a representative human immunodeficiency virus-1+ aviraemic patient (f) in the different T memory subsets.
Levels of PD-L1 expression on CD4 and CD8 T cell subsets
Next, we measured percentages of T cells positive for the expression of PD-L1 (as shown in the histograms in Fig. 3f). We observed an increase in the percentage of PD-L1-expressing cells in all CD4+ T cell subsets, but only the difference on CD4+ TEMRA in aviraemic patients compared to controls reached statistical significance (P = 0·0119) (Fig. 3a). Percentages of PD-L1-expressing cells were not reduced substantially in the viraemic group in all CD4+ T cells subsets when compared to controls. MFI values for PD-L1 were not statistically different in the CD4+ T cells subsets, although some reduction was observed in the naive subsets in both viraemic and aviraemic patients (Fig. 3b).
Fig. 3.
Representation of data for flow cytometric analysis of expression of programmed death ligand (PD-L1) molecule on CD4+[a: percentages; b: mean fluorescence intensity (MFI)] and CD8+ T cells (c: percentages; d: MFI). Bar graphs show mean and error bars represent standard error of the mean in the three different groups: healthy controls (▭), aviraemic patients () and viraemic patients (
). The histograms show PD-L1 expression on the T cell subsets from a representative human immunodeficiency virus-1+ aviraemic patient (f).
Numbers of cells expressing PD-L1 were increased in the CD8+ T cell subsets in the aviraemic HIV-1+group compared to healthy controls, especially in the TEM (P = 0·0494) and the TEMRA subsets (P = 0·0282) (Fig. 3c). A trend towards increased PD-L1-expressing cells was also observed in CD8 TEMRA of viraemic HIV-1+patients. MFI levels of PD-L1 were not statistically different between the three groups in all CD8+ T cell subsets; they appeared to be slightly increased in the healthy control group (Fig. 3d).
PD-1 and PD-L1 blocking
We tested the effect of PD-1 and PD-L1 blockade on HIV-1-specific responses in proliferative and IFN-γ production assays using Gag peptides as stimulus (patients' characteristics are illustrated in Table 1). We found that only one of eight patients responded to Gag in proliferative assays while four of five patients produced IFN-γ (Fig. 4a,b). PD-1 blockade increased Gag-induced proliferation in the responsive patient but failed to rescue responses in any of the other eight patients (Fig. 4a). When we used anti-PD-L1 blocking antibody we measured increased Gag-induced proliferation in the responsive patient and also rescued responses in four more patients who had no response to Gag before PD-L1 blockade (Fig. 4b). In five of the patients, IFN-γ ELISPOT was also performed, but there was no change in IFN-γ production after Gag stimulation either in the absence or presence of PD-1 or PD-L1 blocking antibodies (Fig. 4a,b).
Fig. 4.
Graphs showing stimulation index results and interferon-γ production of human immunodeficiency virus-1+ patients' peripheral blood mononuclear cell stimulation with peptide pools of Gag 20mers and Gag 9mers. The first set of graphs show results using anti-programmed death (PD-1) antibody in lymphocyte proliferative response (LPR) and enzyme-linked immunospot assays (ELISPOT) (a); the last four graphs show LPR and ELISPOT assays using anti-PD-ligand-1 (b).
Discussion
In this study we dissected the phenotypical changes in differentiation and maturation markers affecting CD4+ and CD8+ T cells during HIV-1 infection. It has been reported previously that changes in the distribution of T cells in different memory subsets occur during HIV-1 infection, and may affect effective responses negatively against HIV-1. Our results display a phenotypical picture showing increased numbers of CD4+ and CD8+TEM cells, which are preterminally differentiated producing cytokines but are less capable of dividing in HIV-1+ patients. The increase of TEM seems to be influenced in particular by the presence of virus for the CD8+ T cells. Another important difference observed was the decrease of naive CD8+ T cells in HIV-1 patients, due possibly to impaired haematopoiesis and lack of signalling from the CD4+ T cells [18–20]. We could not find any substantial difference in the TCM subset in CD8+ and CD4+ T cells, which may be explained by the positive effect of HAART in regenerating T cell populations [21,22].
Abnormal distribution between the subsets is accompanied by an increased number of cells expressing PD-1; this phenomenon was particularly evident in the CD4+ T cell compartment, especially in the TEM, TCM and naive T cell subsets in the aviraemic patient group. This observation may explain the lack of help and stimulation by CD4+ T cell to CTL during HIV-1 infection, resulting in an abnormal composition of the CD8+ T cell memory subsets, despite successful HAART [13]. Numbers of cells expressing PD-L1 are increased in the HIV-1+ aviraemic group's CD4+ subsets reinforcing a picture of negative regulation and anergy [8,10]. We found lower numbers of cells expressing PD-L1 in the aviraemic patient group CD4+ T cell subset due probably to the fact that, in this group of patients, cells expressing PD-L1 go into apoptosis more rapidly by inducing production of IFN-α, as reported by Boasso et al.[23].
Interestingly, more naive CD8+ T cells express PD-1 in HIV-1-infected patients than HIV-1− individuals. Also, as reported by other groups, CD8 TEM numbers are increased during HIV-1 infection [13] and express more PD-L1 [10,24,25], which is also increased in the CD8 TEMRA subset.
These results confirm previous findings obtained by other groups using different markers to define CD8 T cell memory subsets [24,25], showing that up-regulation of PD-1 is present not only on the anti-HIV-1-specific CD8 T cell but also in the general CD8 T cell and CD4 T cell populations, despite patients being on successful HAART regimens with suppressed viral load. Previously published results from other groups have also shown the role of PD-1 in CD4 T cell apoptosis during HIV-1 infection [26].
The importance of PD-1 and PD-L1 in affecting T cell functionality, in particular the specific ability to generate responses against HIV-1, is demonstrated by the fact that specific blocking antibodies against the two molecules increased T cell proliferation upon stimulation against Gag.
Blocking PD-1 resulted in increased anti-HIV-1-specific proliferative responses in one of eight patients. It was noted that anti-Gag responses were already present in this patient. A possible explanation for this is the fact that PD-1 expression is normalized by HAART [7].
When PD-L1 was blocked, T cells from four of eight of the HIV-1+ patients were able to proliferate against Gag; only one of the four patients had proliferative responses before the blockade. This concurs with results from other groups [7,9]. The higher efficacy of PD-L1 blockade compared to PD-1 blockade might be explained by the fact that PD-L1 expression is less affected by HAART than PD-1 expression [10]. Furthermore, while anti-PD-1 antibody would be expected to block only the PD-1/PD-L1 interaction, blocking PD-L1 could have a broader stimulatory effect by preventing the binding of PD-L1 to other receptors such as the CD28 receptor B7-1 on both lymphocytes and APC [27]. This leads to the conclusion that, together with HAART, which seems able to restore subset composition (especially in the CD4+ T cell compartment), novel strategies or therapies are needed in order to reverse anergy and rescue the ability of CD8+CTL to respond to HIV-1-specific stimuli. PD-1 and PD-L1 seem to be interesting targets for intervention, bearing in mind that modulation of these molecules will have to be achieved while avoiding the risk of generating autoimmune responses. Longitudinal studies will be required to define more clearly the expression profile of PD-1 and PD-L1 during the course of HIV-1 infection. Further detailed investigations are needed into the interlinked signalling cascade regulating the different co-stimulatory and co-inhibitory molecules and their receptors.
Acknowledgments
We thank Adriano Boasso for helpful discussion and critical review of the manuscript. The authors thank patients and staff at the Chelsea & Westminster Hospital who participated in this study. We are also grateful to the staff of the Clinical Laboratory (Department of Immunology, Imperial College London, Chelsea & Westminster Hospital) for lymphocyte subsets and viral load measurement. This study was supported by funding from AVIP EU Program Grant (no. LSHP-CT-2004-503487). N. I. and F. G. are also funded by the MRC (grant no. G0501957).
Disclosures
The authors have no financial conflict of interest.
References
- 1.Imami N, Hardy G, Burton C, et al. Immune responses and reconstitution in HIV-1 infected individuals: impact of anti-retroviral therapy, cytokines and therapeutic vaccination. Immunol Lett. 2001;79:63–76. doi: 10.1016/s0165-2478(01)00267-x. [DOI] [PubMed] [Google Scholar]
- 2.Valdez H, Connick E, Smith KY, et al. Limited immune restoration after 3 years' suppression of HIV-1 replication in patients with moderately advanced disease. AIDS. 2002;16:1859–66. doi: 10.1097/00002030-200209270-00002. [DOI] [PubMed] [Google Scholar]
- 3.Noel PJ, Boise LH, Green JM, Thompson CB. CD28 costimulation prevents cell death during primary T cell activation. J Immunol. 1996;157:636–42. [PubMed] [Google Scholar]
- 4.Shahinian A, Pfeffer K, Lee KP, et al. Differential T cell costimulatory requirements in CD28-deficient mice. Science. 1993;261:609–12. doi: 10.1126/science.7688139. [DOI] [PubMed] [Google Scholar]
- 5.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–7. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
- 7.Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–4. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
- 8.Trabattoni D, Saresella M, Biasin M, et al. B7-H1 is up-regulated in HIV infection and is a novel surrogate marker of disease progression. Blood. 2003;101:2514–20. doi: 10.1182/blood-2002-10-3065. [DOI] [PubMed] [Google Scholar]
- 9.Trautmann L, Janbazian L, Chomont N, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–202. doi: 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
- 10.Rosignoli G, Cranage A, Burton C, et al. Expression of PD-L1, a marker of disease status, is not reduced by HAART in aviraemic patients. AIDS. 2007;21:1379–81. doi: 10.1097/QAD.0b013e3281de7296. [DOI] [PubMed] [Google Scholar]
- 11.Douek DC, Brenchley JM, Betts MR, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature. 2002;417:95–8. doi: 10.1038/417095a. [DOI] [PubMed] [Google Scholar]
- 12.Grossman Z, Meier-Schellersheim M, Paul WE, Picker LJ. Pathogenesis of HIV infection: what the virus spares is as important as what it destroys. Nat Med. 2006;12:289–95. doi: 10.1038/nm1380. [DOI] [PubMed] [Google Scholar]
- 13.Champagne P, Ogg GS, King AS, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature. 2001;410:106–11. doi: 10.1038/35065118. [DOI] [PubMed] [Google Scholar]
- 14.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–12. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
- 15.Harari A, Petitpierre S, Vallelian F, Pantaleo G. Skewed representation of functionally distinct populations of virus-specific CD4 T cells in HIV-1-infected subjects with progressive disease: changes after antiretroviral therapy. Blood. 2004;103:966–72. doi: 10.1182/blood-2003-04-1203. [DOI] [PubMed] [Google Scholar]
- 16.Hardy GA, Imami N, Nelson MR, et al. A phase I, randomized study of combined IL-2 and therapeutic immunisation with antiretroviral therapy. J Immune Based Ther Vaccines. 2007;5:6. doi: 10.1186/1476-8518-5-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Imami N, Pires A, Hardy G, Wilson J, Gazzard B, Gotch F. A balanced type 1/type 2 response is associated with long-term nonprogressive human immunodeficiency virus type 1 infection. J Virol. 2002;76:9011–23. doi: 10.1128/JVI.76.18.9011-9023.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kryworuchko M, Pasquier V, Theze J. Human immunodeficiency virus-1 envelope glycoproteins and anti-CD4 antibodies inhibit interleukin-2-induced Jak/STAT signalling in human CD4 T lymphocytes. Clin Exp Immunol. 2003;131:422–7. doi: 10.1046/j.1365-2249.2003.02065.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science. 1997;278:1447–50. doi: 10.1126/science.278.5342.1447. [DOI] [PubMed] [Google Scholar]
- 20.Elrefaei M, McElroy MD, Preas CP, et al. Central memory CD4+ T cell responses in chronic HIV infection are not restored by antiretroviral therapy. J Immunol. 2004;173:2184–9. doi: 10.4049/jimmunol.173.3.2184. [DOI] [PubMed] [Google Scholar]
- 21.Haase AT. Population biology of HIV-1 infection: viral and CD4+ T cell demographics and dynamics in lymphatic tissues. Annu Rev Immunol. 1999;17:625–56. doi: 10.1146/annurev.immunol.17.1.625. [DOI] [PubMed] [Google Scholar]
- 22.Burton CT, Mela CM, Rosignoli G, Westrop SJ, Gotch FM, Imami N. Immune modulation and reconstitution of HIV-1-specific responses: novel approaches and strategies. Curr Med Chem. 2006;13:3203–11. doi: 10.2174/092986706778742972. [DOI] [PubMed] [Google Scholar]
- 23.Boasso A, Hardy AW, Landay AL, et al. PDL-1 upregulation on monocytes and T cells by HIV via type I interferon: restricted expression of type I interferon receptor by CCR5-expressing leukocytes. Clin Immunol. 2008;129:132–44. doi: 10.1016/j.clim.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sauce D, Almeida JR, Larsen M, et al. PD-1 expression on human CD8 T cells depends on both state of differentiation and activation status. AIDS. 2007;21:2005–13. doi: 10.1097/QAD.0b013e3282eee548. [DOI] [PubMed] [Google Scholar]
- 25.Petrovas C, Casazza JP, Brenchley JM, et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J Exp Med. 2006;203:2281–92. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Venkatachari NJ, Buchanan WG, Ayyavoo V. Human immunodeficiency virus (HIV-1) infection selectively downregulates PD-1 expression in infected cells and protects the cells from early apoptosis in vitro and in vivo. Virology. 2008;376:140–53. doi: 10.1016/j.virol.2008.03.015. [DOI] [PubMed] [Google Scholar]
- 27.Butte MJ, Pena-Cruz V, Kim MJ, Freeman GJ, Sharpe AH. Interaction of human PD-L1 and B7-1. Mol Immunol. 2008;45:3567–72. doi: 10.1016/j.molimm.2008.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]