Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 1.
Published in final edited form as: J Immunol. 2013 Aug 5;191(5):10.4049/jimmunol.1200646. doi: 10.4049/jimmunol.1200646

PD-1 is a marker for abnormal distribution of naïve/memory T cell subsets in HIV-1 infection

Gaëlle Breton 1,2,3, Nicolas Chomont 1,2,3,4, Hiroshi Takata 4, Rémi Fromentin 4, Jeffrey Ahlers 4, Abdelali Filali-Mouhim 4, Catherine Riou 1,2,3, Mohamed-Rachid Boulassel 5, Jean-Pierre Routy 5,6, Bader Yassine-Diab 1,2,3, Rafick-Pierre Sékaly 1,2,3,4,6
PMCID: PMC3815464  NIHMSID: NIHMS500319  PMID: 23918986

Abstract

Chronic activation of T cells is a hallmark of HIV-1 infection and plays an important role in disease progression. We previously showed that the engagement of the inhibitory receptor PD-1 on HIV-1 specific CD4+ and CD8+ T cells leads to their functional exhaustion in vitro. However, little is known about the impact of PD-1 expression on the turnover and maturation status of T cells during the course of the disease. Here, we show that PD-1 is up regulated on all T cell subsets including naïve, central memory and transitional memory T cells in HIV-1 infected subjects. PD-1 is expressed at similar levels on most CD4+ T cells during the acute and the chronic phase of disease and identifies cells that have recently entered the cell cycle. In contrast, PD-1 expression dramatically increased in CD8+ T cells during the transition from acute to chronic infection and this was associated with reduced levels of cell proliferation. The failure to down regulate expression of PD-1 in most T cells during chronic HIV-1 infection was associated to persistent alterations in the distribution of T cell subsets and was associated with impaired responses to IL-7. Our findings identify PD-1 as a marker for aberrant distribution of T cell subsets in HIV-1 infection.

Keywords: HIV-1, PD-1, CD8+ and CD4+ T cell, homeostasis, Acute and chronic infection

Introduction

Acute HIV-1 infection leads to the massive depletion of mucosal CD4+ T cells and progressive dysfunction of cellular and humoral immune responses in the majority of infected individuals. Rapid replication of virus in CD4+ T cells, including Th17 cells in mucosal lymphoid tissue, concomitant with inflammatory cytokine and chemokine production leads to disruption of the gut mucosal barrier permitting commensal bacteria to enter into the blood stream resulting in systemic activation of innate immune cells by microbial products (1).

Chronic activation of the immune system is characterized by (i) high levels of expression of activation markers including HLA-DR, CD38, and the nuclear antigen Ki67 on both CD4+ and CD8+ T cells (26) (ii) increased susceptibility to activation induced cell death (7) (iii) increased plasma levels of inflammatory cytokines and Type I IFNs (8) (iv) accumulation of terminally differentiated CD4+ and CD8+ T cells as shown by the loss of expression of the CD28 co-stimulatory molecule and increased levels of CD57 expression (911). Chronic immune activation is now recognized as the sine qua non for HIV-1 pathogenesis and is a better predictor of HIV-1 disease progression than the extent of viral replication (6, 1214). Cross-sectional studies of immune function in HIV-1 infected individuals including global gene expression analyses of innate and adaptive cell populations suggest that the host immune response cannot contain virus replication and that the cumulative effects of Type I IFNs, proinflammatory and profibrinogenic cytokines can explain, in large part, the ensuing global defects in T and B cell function, homeostasis, and cell death (1525).

The PD-1/PD-L1 co-inhibitory pathway plays a significant role in the regulation of the immune response and dysfunction in chronic infectious diseases and cancer (26). The increased levels of expression of Programmed death-1 (PD-1) molecules on antigen-specific T cells in persistent infection, including HIV-1 specific T cells, provides a signature of functional T cell exhaustion. PD-1, a T cell activation marker, is up regulated upon TCR triggering and accumulates at the immunological synapse (27). Upon engagement by its ligand PD-L1 or PD-L2, PD-1 signaling enforces an inhibitory program that blocks further TCR-induced T cell proliferation and cytokine production (28, 29). In a typical acute viral infection this mechanism constitutes a feedback loop that limits the increase in absolute numbers of effector cells during the expansion phase of the immune response. However, under conditions of chronic antigen exposure, this pathway impairs the responsiveness of T cells ultimately leading to the exhaustion of these cells. Studies from a number of groups have shown that during chronic HIV-1 infection PD-1 expression on HIV-1 specific T cells correlates with viral load and that blocking PD-1 engagement restores T-cell effector functions in vitro (3034). In addition, in vivo PD-1 blockade in chronic SIV infection restored CD8+ T cell function, reduced viral load levels and increased survival of SIV infected macaques (35).

While the role of PD-1 in T cell exhaustion during chronic HIV-1 infection is well established, its function during acute HIV-1 infection is much less clear. Therefore we performed a cross-sectional analysis of PD-1 expression in CD4+ and CD8+ T cell subsets in a total of 51 individuals at all stages of disease, including individuals receiving HAART and uninfected donors.

Materials and methods

Study population

For phenotypic characterization, 13 subjects with chronic HIV-1 infection (CHI), 8 individuals with acute HIV-1 infection (AHI), 15 subjects receiving highly active antiretroviral therapy (HAART) and 15 HIV-1 seronegative control individuals were enrolled. A summary of these patients clinical data is shown in Table I. To assess the capacity of memory T cells to respond to IL-7 stimulation, 8 subjects with chronic HIV-1 infection (CHI) and 5 HIV-1 seronegative control individuals were enrolled. A summary of these patients clinical data is shown in Table II. None of the HIV-1 acute or chronic infected patients were on antiretroviral therapy at the time of this study. The following guidelines proposed by the Acute HIV Infection and Early Disease Research Program sponsored by the National Institutes of Allergy and Infectious Disease Division of AIDS (Bethesda, Maryland) were used to estimate the date of infection: 1) the date of the first positive HIV RNA test or p24 Ag assay available on the same day as a negative standard HIV enzyme immunoassay test minus 14 days; 2) the date of onset of symptoms of an acute retroviral syndrome minus 14 days; 3) the date of the first indeterminate Western blot minus 35 days; 4) the detuned assay (as described in reference (36)). All patients signed informed consent approved by the Royal Victoria Hospital and CR-CHUM review boards.

Table I.

Clinical characteristics of HIV-1 infected subjects

Patient Viral loada CD4 countb CD8 countc Treatmentd Time of infectione
Chronic-untreated
VR DRPI 025 218113 296 364 untreated 24 months
VR 003001 200363 310 350 untreated 32 months
VR 378 176557 443 736 untreated 5 months
VR 021 97044 730 1310 untreated 5 months
VR 42117 68412 704 1081 untreated 8 months
VR 360 57154 316 376 untreated 8 months
VR 026 36349 442 538 untreated 6 months
VR 025 20840 479 791 untreated 7 months
VR 003 15703 494 1055 untreated 16 months
VR 016 8714 691 1122 untreated 12 months
VR 347 4456 1173 1143 untreated 10 months
VR 03006 2021 522 366 untreated 5 months
VR 026 1705 368 425 untreated 12 months
Median 28594 487 763

Acute-untreated
ACU 365 3221835 257 1007 untreated 43 days
ACU 013 366646 483 930 untreated 51 days
ACU 011 125536 594 692 untreated 52 days
ACU 375 93706 378 779 untreated 72 days
ACU DRPI 039 93223 857 1499 untreated 91 days
ACU 373 81984 338 1829 untreated 106 days
ACU 360 59925 359 706 untreated 87 days
ACU DRPI 041 56838 475 640 untreated 57 days
Median 93706 439 930

Chronic-treated
ST 101002 <50 662 1051 DDI D4T EFV 19 months
ST 008 <50 463 376 NFV D4T 3TC 20 months
ST 310 <50 650 392 D4T 3TC IND 64 months
ST 325 <50 604 1281 IND COM 53 months
ST 326 <50 883 333 EFV IND 49 months
ST 338 <50 443 322 RIT COM KAL 18 months
ST 001 <50 890 673 COM NEV 57 months
ST 002 <50 688 1273 COM NEV 100 months
ST 003 <50 434 583 3TC EFV ABC 165 months
ST 004 <50 492 582 RIT REY KIV 166 months
ST 007 <50 563 613 IND AZT 3TC 86 months
ST 009 <50 424 461 3TC D4T DEL 84 months
ST 015 <50 552 715 D4T ATA 139 months
ST 016 <50 671 1120 3TC ABC KAL 242 months
ST 017 <50 510 765 COM RIT 61 months
Median <50 563 613
a

Copies of HIV-1 RNA/ml of plasma at the time of study

b

Peripheral CD4+ T lymphocytes count at the time of study

c

Peripheral CD8+ T lymphocytes count at the time of study

d

Antiretroviral drug abbreviations: ABC, abacavir; ATA, atazanavir; AZT, zidovudine; COM, combivir; DDI, didanosine; DEL, delavirdine; D4T, stavudine; EFV, efavirenz; IND, indinavir; KAL, kaletra; KIV, kivexa; NFV, nelfinavir; NEV, nevirapine; REY, reyataz; RIT, ritonavir; 3TC, lamivudine.

e

Duration of infection at the time of study

Table II.

Clinical characteristics of HIV-1 infected subjects

Patient Viral loada CD4 countb CD8 countc Treatmentd Time of infectione
Chronic-untreated
VR 404 51000 434 2497 untreated 30 months
VR 405 23000 548 751 untreated 224 months
VR 406 5700 504 958 untreated 327 months
VR 407 3300 480 1552 untreated 68 months
VR 408 400000 258 1704 untreated 62 months
VR 409 50000 185 647 untreated 115 months
VR 410 21000 284 605 untreated 68 months
VR 001 52915 499 1590 untreated 120 months
Median 75864 399 1288

Preparation of PBMCs

Peripheral blood mononuclear cells (PBMCs) were isolated from heparinized blood by density gradient centrifugation (Ficoll-Paque) and cryopreserved in liquid nitrogen.

Antibodies and reagents

For phenotypic characterization, the following monoclonal antibodies (mAbs) were used: anti-CD3-A700 (UCHT1), anti-CD4-PB (RPA-T4), anti-CD8-PE-TR (MHCD0817), anti-CD28-PE-Cy5 (CD28.2), anti-CD45RA-APC (HI100), anti-CCR7-PE-Cy7 (3D12), anti-CD27-APC-Cy7 (M-T271), anti-PD-1-PE (MIH4), anti-CD57-FITC (HNK-1) and anti-Ki67-FITC (B56). For phosflow analysis, the following mAbs were used: anti-CD3-PE-Cy7 (UCHT1), anti-CD4-PerCP-Cy5.5 (RPA-T4), anti-CD8-PB (RPA-T8), anti-CD45RA-ECD (2H4LDH11LDB9), anti-CCR7-FITC (150503), anti-CD27-Qdot655 (CLB-27/1), anti-PD-1-PE (J105), Live/dead Aqua and anti-Stat5a-Alexa647 (41/Stat5(pY694)). For Bcl-2 intracellular staining, the following antibodies were used: anti-CD3-A700 (UCHT1), anti-CD4-Qdot605 (S3.5), anti-CD8-PB (RPA-T8), anti-CD45RA-BV650 (HI100), anti-CCR7-PE-Cy7 (3D12), anti-CD27-APC-H7 (M-T271), anti-PD-1-PE (J105), Live/dead Aqua and anti-Bcl-2-FITC (Bcl-2/100). Recombinant human IL-7 was purchased from R&D Systems.

Phenotypic characterization by flow cytometry

Cells from all donors were simultaneously stained with two 9-color 11-parameter staining cocktails (Cocktail 1: CD3, CD4, CD8, CD45RA, CCR7, CD27, CD28, PD-1 and CD57; cocktail 2: CD3, CD4, CD8, CD45RA, CCR7, CD27, CD28, PD-1 and Ki67). FACS staining was performed on ex-vivo whole PBMC. Titrated antibodies were added to 2 million cells in 50μl of PBS for 20 minutes at 4°C. Intracellular staining for Ki67 was conducted by incubating cells stained for cell surface markers with FACS permeabilizing solution (BD Biosciences) for 20 minutes at room temperature. Cells were then stained in permeabilization buffer (PBS containing 0.05% saponin) with Ki67 antibody or isotype control for 30 minutes at room temperature. Washed cells were fixed in 2% formaldehyde and stored at 4°C until analysis, which was performed using a LSR II flow cytometer (BD Biosciences). The whole sample was acquired and the lymphocytes were gated for further analysis, as described in Fig. 1A, using Diva 4.1 software (BD Biosciences).

Figure 1.

Figure 1

Disruption of T cell subsets homeostasis in HIV-1 infection is specific to each disease stage. A. Seven-parameter flow cytometry gating strategy to identify CD4+ and CD8+ T cell subsets. TN, TCM, TTM, TEM and TTD/TE subsets are identified based on their CD45RA, CCR7 and CD27 expression. B. Distribution of blood CD4+ and CD8+ T cell subpopulations in acute-untreated (n=8; black symbol), chronic-untreated (n=13; red symbol), chronic-treated (n=15; blue symbol) and uninfected (n=15; green symbol) subjects. Each symbol represents one subject. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

Phosflow analysis of Stat5a phosphorylation

PBMCs were first stained with Live/dead, anti-CD27, anti-CCR7, and anti-PD-1 mAbs for 30 min at 4°C. The cells were washed with 2% FCS PBS (staining buffer) and resuspended at 20 million cells/ml in RPMI 1640 and stimulated for 15 min at 37°C in the presence of 0.3 ng/ml IL-7. After stimulation, the cells were fixed for 10 min at 37°C with PBS 2% formaldehyde, pelleted, and permeabilized in PERM III buffer (BD Biosciences) for 30 min on ice. The cells were washed twice in staining buffer and rehydrated for 30 min at 4°C in the staining buffer. The cells were stained with anti-CD3, anti-CD4, anti-CD8, anti-CD45RA, and anti-pStat5a (Y694) mAbs for 30 min at room temperature. Cells were acquired on a LSR II flow cytometer (BD Biosciences) and flow cytometry data were analyzed using FlowJo 8.9 software (TreeStar).

Bcl-2 intracellular staining

PBMCs were first labeled with surface markers for 30 min at 4°C. The cells were washed twice with PBS 2% FCS and fixed in 2% formaldehyde for 20 min at room temperature. The cells were permeabilized in PBS containing 0.1% saponin and 10% FCS for 10 min at 4°C and then stained with anti-Bcl-2 for 30 min at room temperature. Finally, cells were washed and fixed in 2% formaldehyde. For the analysis, cells were acquired on a LSR II flow cytometer (BD Biosciences) and the isotype matched antibody was used as negative control. All flow cytometry data were analyzed using FlowJo 8.9 software (TreeStar).

Statistical analysis

Correlations were performed using the Spearman test and statistical significances between groups were calculated by Mann-Whitney U test using Prism 3.0 software (GraphPad). We used the Receiver Operating Characteristic (ROC) and the area under the ROC curve (AUC) to evaluate the performance of the Fisher’s Linear Discriminant analysis (LDA) classifier to predict the HIV status (acute/chronic) in one and two variables models using Leaving-One-Out Cross Validation (LOOCV) method. We generated Receiver Operating Characteristic (ROC) curves by calculating the classifier’s true positive rate (sensitivity) and false positive rate (100-Specificity) at various class membership probabilities scores yielded by the classifier. Each value of the score yields a single point on the ROC curve. The area under the ROC curve provides a measure of how well the predictor variables tested could distinguish between two groups (acute/chronic). The class prediction analysis was performed using CMA, a package from Bioconductor (www.bioconductor.org), an open-source software library for the analysis of genomic data based on R, a language and environment for statistical computing and graphics (www.r-project.org).

Results

HIV-1 infection is associated with abnormal distribution of T cell subsets specific to each stage of the disease and incompletely restored by HAART

To study the impact of HIV-1 infection on the distribution of T cell subsets, we first measured the frequencies of CD4+ and CD8+ T cell sub-populations in individuals at 3 different stages of the disease (Table I). The combined use of the CD45RA, CCR7 and CD27 markers allowed us to identify five phenotypically and functionally distinct populations in both CD4 and CD8 compartments: Naïve (TN: CD45RA+, CD27+, CCR7+), Central memory (TCM: CD45RA, CD27+, CCR7+), Transitional memory (TTM: CD45RA, CD27+, CCR7), Effector memory (TEM: CD45RA, CD27, CCR7) and Effector CD8+ T cells and Terminally differentiated CD4+ T cells (TE and TTD; CD45RA+ CD27, CCR7) (Fig. 1A).

Acute HIV-1 infection (AHI) was characterized by increased frequencies of CD8+ TTM and TEM cells (p<0.0001 and p<0.05 respectively) concomitant with decreased frequencies of CD8+ TN and TCM cells (p<0.0001 and p<0.001 respectively) when compared to control donors (Fig. 1B). Remarkably, the CD8+ TTM cells expansion during AHI is dramatic (2.3 fold increase). In addition, increased frequencies of CD4+ TEM cells (p<0.001) were also noted in acutely infected subjects when compared to uninfected individuals (Fig. 1B). This shift towards a more differentiated T cell phenotype observed in AHI likely reflects strong priming of naïve T cells and their differentiation into a large population of end stage effector cells, a hallmark of acute viral infections (37). Although CD4+ TEM and CD8+ TEM cells were found at high frequencies in both AHI and chronic HIV-1 infection (CHI) subjects, the transition from the acute to the chronic phase was accompanied by an increased frequency of TN in both compartments (p<0.05 for the CD4 compartment and p<0.001 for the CD8 compartment), while the frequencies of CD4+ TCM, CD4+ TTM and CD8+ TTM decreased (p<0.05, p<0.05 and p<0.0001 respectively) (Fig. 1B). These results confirm previous findings (24) that showed a more differentiated phenotype of CD4+ and CD8+ T cells in chronically infected subjects when compared to HIV-1 negative donors.

A similar analysis performed in aviremic treated individuals showed that HAART was able to restore the distribution of all memory CD4+ T cell subsets to the frequencies observed in healthy donors. However, HAART failed to restore normal CD8+ T cell subset frequencies, with persisting disequilibrium shown by lower numbers of TCM (p<0.05) and higher numbers of TEM (p<0.05) subsets compared to uninfected subjects (Fig. 1B). Thus, despite effective antiretroviral treatment, even for a prolonged period of time, the CD8+ TCM and TEM compartments are not fully restored to the homeostatic levels found in uninfected controls.

Altogether, our results indicate that HIV-1 infection leads to a disequilibrium in the frequency of CD4+ and CD8+ T cell subsets in both acutely and chronically infected patients. The distribution of most if not all T cell subsets in both CD4 and CD8 compartments are affected. More importantly, differences observed are specific to each stage of disease and response to HAART.

HIV-1 infection is associated with altered differentiation of T cell subsets and senescence

To characterize the extent of differentiated T cell populations in HIV-1 infected individuals and controls we assessed the relative levels of expression of CD28 and CD57 on both CD4+ and CD8+ T cell subsets. A number of groups have shown that the loss of CD28 expression as well as increased levels of CD57 expression are associated with a state of replicative senescence that correlates with HIV-1 disease progression (9, 38, 39). As shown in Fig. 2A, loss of CD28 expression was observed in uninfected individuals in the late differentiated TEM and TTD subsets in the CD4 compartment whereas CD28 expression in the CD8 compartment progressively decreased with differentiation to ~43% of total effector cells.

Figure 2.

Figure 2

Differentiation toward senescent stages of all T cell subsets characterized by loss of CD28 expression and increased CD57 expression in HIV-1 infection. a. Percentage of ex-vivo CD28 expressing cells in CD4+ and CD8+ T cell subsets in HIV-1 infected individuals and controls. b. Percentage of ex-vivo CD57 expressing cells in CD4+ and CD8+ T cell subsets in HIV-1 infected individuals and controls. Each symbol represents one subject: acute-untreated (n=8; black symbol), chronic-untreated (n=13; red symbol), chronic-treated (n=15; blue symbol) and uninfected (n=15; green symbol) subjects. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

The percentage of CD8+ T cell expressing CD28 was most significantly reduced in the AHI individuals compared to uninfected individuals (TCM, TTM, TEM p<0.0001 and for TE p<0.001). Reduced frequencies of CD28+ CD8+ TN, TCM and TTM subsets were also more pronounced during AHI than during CHI (p<0.05 for all subsets). Importantly, we found a significant reduction in the frequency of CD8+ TCM that expressed CD28 in both acute and chronic infection, which is consistent with the lower frequency of cells in this subset. When compared to the control group, CHI and AHI subjects showed significantly lower percentages of CD4+ CD28+ cells. Chronic infected individuals showed significant reduction in the frequencies of CD4 cells expressing CD28 in activated TTM, TEM and TTD subsets (p<0.001) as well as TCM subset (p<0.05) compared to untreated controls, whereas the percentage of CD4+ CD28+ cells was similar between the acutely and chronically infected individual. Lastly, the frequencies of CD8+ CD28+ cells in HAART treated subjects never fully recovered to levels observed in uninfected individuals (TTM, p<0.001; TEM and TE, p<0.05) whereas the only significant differences in CD4+ CD28+ expression between HAART-treated and uninfected occurred in the effector memory pool (TEM, p<0.001).

Since CD57 expression has been associated with replicative senescence and apoptosis in HIV infected individuals (40), we next assessed the percentage of T cells that expressed CD57 in each of the CD4 and CD8 subpopulations (Fig. 2B). As expected, the up regulation of CD57 expression occurred mainly in the most differentiated TEM and TTD/E subsets in both CD4+ and CD8+ T cells and exhibited a wide range of individual responses within each group.

With the exception of CD4+ TN cells, all CD4+ subsets from CHI subjects showed an increased percentage of CD57+ cells when compared to controls (TCM, p<0.05; TTM, TEM, TTD, p<0.001). All CD8+ subsets also displayed increased percentage of CD57+ cells (TN, p<0.05; TCM p<0.0001; TTM, TEM and TE, p<0.05). It appears that T cells start to express CD57 very early in acute HIV-1 infection, as all the subsets show significantly increased numbers of CD57+ cells compared to uninfected subjects with: CD4+ TCM (p<0.05), CD4+ TTM (p<0.001), CD4+ TEM (p<0.001), CD4+ TTD (p<0.001), CD8+ TN (p<0.001), CD8+ TCM (p<0.0001), CD8+ TTM (p<0.05) and CD8+ TE (p<0.05). It is important to note that we did not see any significant differences in CD57 expression between CHI and AHI. Virally suppressed subjects also displayed abnormally high frequencies of CD57+ cells in the CD4+ TEM (p<0.05), CD8+ TCM (p<0.05) and CD8+ TE (p<0.05) subsets compared to uninfected controls, once again indicating that HAART does not completely restore the replicative capability and clonal turnover of effector and effector memory T cell populations. Consistent with this hypothesis, we found a strong inverse correlation between the percentage of CD57+ cells and the percentage of CD28+ cells in individual CD4+ and CD8+ naïve/memory subsets, suggesting that the loss of CD28 expression was accompanied by the up regulation of the CD57 marker (Data not shown).

Altogether our results demonstrate that during HIV-1 infection CD28 cells and CD57+ cells can be found in most CD4+ and CD8+ subsets in addition to the terminally differentiated senescent cell population usually described in chronically infected individuals. Markers associated with senescence were mainly attributed to memory subsets in the CD4 compartment and found in all subsets in the CD8 compartment. Importantly, we found no significant differences in the frequencies of T cell subsets expressing either CD28 or CD57 between CHI and AHI indicating that functional impairment of T cell responses reaches a plateau within 1–3 months after infection (Table I).

T cell proliferation in HIV-1 infected individuals is dependent on the disease stage and is incompletely restored by HAART

To better assess the extent to which HIV-1 infection drives CD4+ and CD8+ T cell replicative senescence, we measured the fraction of proliferating (i.e. Ki67+) cells in all T cell subsets from all subjects enrolled in this study (Supplemental Fig. 1A). Ki67 is a commonly used and reliable marker of T cell turnover, thereby reflecting the level of immune activation in HIV-1 pathogenesis (26).

In untreated HIV-1 infected subjects, irrespective of the disease status (acute or chronic), all T cell subsets showed significantly higher frequencies of Ki67+ cells when compared to uninfected controls (p<0.0001 for all CD4+ and CD8+ subsets in AHI and CHI) (Fig. 3). Interestingly, even TN cells expressed higher levels of Ki67 in both chronically and acutely HIV-1 infected subjects compared with uninfected controls (p<0.0001). The increased percentages of CD4+ Ki67+ TN in acute infection yet unaltered expression of CD28 and CD57 suggests that cell death and lymphopenia is perturbing normal homeostatic proliferation at the earliest stages of HIV-1 infection.

Figure 3.

Figure 3

Increased percentage of Ki67+ proliferating cells in CD4+ and CD8+ T cell subsets during HIV-1 infection. Bars represent mean values of acute-untreated (n=8; black bar), chronic-untreated (n=13; red bar) chronic-treated (n=15; blue bar) and uninfected (n=15; green bar) subjects. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

CD8+ T cell subsets during AHI also showed higher frequencies of Ki67+ cells compared to CHI (CD8+ TN and TCM, p<0.001; for CD8+ TTM and TEM p<0.05) in contrast to similar frequencies of CD4+ Ki67+ T cell throughout infection. This observation can be explained by high levels of virus replication in acute infection driving CD8+ T cell activation and expansion that occurs in response to virus replication and the emergence of escape variants. When compared to uninfected controls, frequencies of Ki67+ cells remained significantly higher in all CD4+ and CD8+ T cell subsets with the exception of CD8+ TN in subjects receiving suppressive HAART (p<0.05 for CD4+ TN, p<0.001 for CD4+ TCM, p<0.0001 for CD4+ TTM, TEM, and TTD cells; p<0.001 for CD8+ TCM and TTM, p<0.0001 for CD8+ TEM and TE cells) suggesting that residual viremia in treated subjects could induce low levels of CD8+ T cell proliferation and CD4+ T cell homeostatic proliferation in response to persistent CD4+ T cell depletion.

All T cell subsets in both acute and chronic HIV-1 infections show increased PD-1 expression

To investigate whether the expression of the co-inhibitory receptor PD-1 on T cells correlated with perturbations in CD4+ T and CD8+ T cell subsets that we observed throughout HIV-1 infection, we analyzed levels of PD-1 expression on all T cell subsets in our different cohorts (Supplemental Fig. 1B). We observed significantly higher levels of PD-1 expression on total CD4+ and CD8+ T cells in both acutely (p<0.0001 for both subsets) and chronically (p<0.0001 for CD4+ T cells and p<0.001 for CD8+ T cells) infected subjects when compared to uninfected donors (Supplemental Fig. 2A). Interestingly, levels of PD-1 expression on CD4+ T cells were not statistically different between acute and chronic infection whereas PD-1 levels significantly increased on CD8+ T cells during chronic infection (p<0.05). The initiation of HAART led to a marked decrease in levels of PD-1 expression in both CD4 and CD8 compartments (fold decrease = 1.6 and 2.0 respectively, Supplemental Fig. 2A).

AHI was characterized by a marked increase in the levels of PD-1 expression in all CD4+ T cell subsets (p<0.0001 for TN, TCM, TTM, TTD and p<0.001 for TEM subsets; range in fold increase = 1.6–2.0) while this up regulation was found exclusively in TN (p<0.001), TCM (p<0.001) and TTM (p<0.05) subsets in the CD8 compartment (range in fold increase = 1.3–1.9). In contrast, the transition from AHI to CHI was accompanied by significantly increased levels of PD-1 expression in all CD8+ T cell subsets (p<0.05 for TN, TEM and TE subsets; p<0.001 for TCM subset; p<0.0001 for TTM; range in fold increase = 1.4–1.7); the levels of PD-1 expression were even further augmented in the three CD8+ T cell subsets that already expressed PD-1 at high levels during acute infection. On the other hand, PD-1 expression levels were comparable in AHI and CHI in all CD4+ T cell subsets (with the exception of the CD4+ TN subset; p<0.05) (Fig. 4A). Importantly, we observed a positive correlation between PD-1 expression and viral load in the CD4 compartment in CHI but not in AHI, suggesting differential association between PD-1 and viral load in AHI and CHI subjects (Supplemental Fig. 3). Although our group and others reported a correlation between viral load and PD-1 expression in the CD8 compartment in CHI in previous studies (30, 31), we did not observe such a correlation in the present study, most probably due to the limited number of subjects included.

Figure 4.

Figure 4

PD-1 up regulation on all T cell subsets in both acute and chronic infection. Each symbol represents one subject: acute-untreated (n=8; black symbol), chronic-untreated (n=13; red symbol), chronic-treated (n=15; blue symbol) and uninfected (n=15; green symbol) subjects. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

To investigate the effectiveness of HAART to reduce levels of PD-1 expression in CD4+ and CD8+ T cells subsets, we then compared the expression levels of PD-1 in T cells from subjects receiving suppressive HAART and uninfected controls. Our results showed that PD-1 remained up regulated in the late differentiated CD4 subsets (p<0.05 for TEM and p<0.001 for TTD) and in the less differentiated CD8 subsets (p<0.001 for TN and p<0.05 for TCM) even after prolonged antiviral treatment (Fig. 4A).

Taken together, our results show that (i) HIV-1 infected patients express higher level of PD-1 than uninfected individuals in all naïve/memory subsets with the exception of late differentiated CD8+ TEM and TE subsets (ii) PD-1 expression is higher in all CD8 subsets during CHI when compared to AHI and (iii) HAART treatment can restore PD-1 expression to the levels observed in uninfected donors in most but not all CD4 and CD8 subsets.

Expression of PD-1 is associated with a reduced ability of CD8+ T cells to proliferate in chronic but not in acute HIV-1 infection

Since we showed that PD-1 was associated with a skewed distribution of naïve and memory CD8+ T cells subsets during the course of the disease, we extended our analysis to determine the relationship between PD-1 up regulation and T cell turnover as monitored by the expression of Ki67. We focused our analysis on CD8+ TN, TCM, TTM and TEM subsets that showed reduced levels of Ki67 associated with proliferative capability during the chronic stage of infection (Fig. 3). We found that the reduction in proliferative potential was accompanied by an increased expression of PD-1 (Fig. 4). Interestingly, when examining Ki67 and PD-1 coexpression, AHI and CHI subjects segregated from each other, with the acute infected cohort expressing both PD-1 and Ki67 whereas the chronically infected subjects expressed only PD1 (Fig. 5A). Using a class prediction algorithm to predict HIV disease status (acute/chronic), we observed that combining Ki67 and PD-1 improves the prediction performance in comparison to Ki67 and PD-1 tested individually (Supplemental Table I), and allowed us to discriminate acute from chronic HIV-1 infection with high confidence. Thus, the expression of low levels of PD-1 expression together with high frequencies of Ki67+ on CD8 TCM was the best predictor of acute infection (AUC = 0.98). Importantly, a similar analysis performed on CD4+ T cells could not distinguish AHI and CHI subjects (data not shown), suggesting that this phenotype of PD-1low Ki67+ was a unique feature of functional CD8+ T cells during acute infection whereas the phenotype PD-1high Ki67 identified a population of exhausted CD8+ T cells.

Figure 5.

Figure 5

A. Concomitant expressions of Ki67 and PD-1 in CD8+ T cell subsets allow to discriminate between acute and chronic HIV-1 infection. Each symbol represents one subject: acute-untreated (n=8; black symbol), chronic-untreated (n=13; red symbol), chronic-treated (n=15; blue symbol) and uninfected (n=15; green symbol) subjects. B. Expression of PD-1 on Ki67+ cells versus Ki67 cells in the CD8 compartment is much higher in chronic than in acute HIV-1 infection. Each bar represents one group of subjects: acute-untreated (n=8; black bar), chronic-untreated (n=13; red bar), chronic-treated (n=15; blue bar) and uninfected (n=15; green bar) subjects. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

To further delineate between PD-1 as an activation marker and PD-1 as an exhaustion marker, we assessed the levels of expression of PD-1 on Ki67+ versus Ki67 cells in total CD4+ and CD8+ T cells (Fig. 5B). Ki67+ and Ki67 CD4+ T cells from control donors as well as successfully treated subjects expressed similar levels of PD-1. On the other hand, CD4+ Ki67+ T cells from both chronically (p<0.0001) and acutely (p<0.001) HIV-1 infected individuals expressed significantly higher levels of PD-1 than their Ki67 counterparts, suggesting that PD-1 expression reflects the continuous proliferation and differentiation of CD4+ T cells during acute and chronic HIV-1 infection. Moreover, Ki67+ PD-1high CD4+ T cells were found at similar frequencies during AHI (3.5 %) and CHI (2.8 %) (Supplemental Fig. 2B) in spite of the different distributions of CD4+ T cell subsets observed between the two disease stages (Fig. 1B). This observation indicates that a small pool (2–4 %) of proliferating PD-1high Ki67+ CD4+ T cells are present at constant frequencies throughout the disease progression.

We performed a similar analysis on total CD8+ T cells from HIV-1 infected individuals and observed a completely different pattern. Indeed, both proliferating and non-proliferating CD8+ T cells expressed similar or lower levels of PD-1 during acute infection. In contrast, proliferating cells from chronically infected subjects showed higher levels of PD-1 when compared to non-proliferating CD8+ T cells (p<0.0001) and this was accompanied by a dramatic decrease in the frequency of proliferating CD8+ T cells when compared to AHI (from 16% to 8%; p<0.001) (Supplemental Fig. 2B). This suggests that the high levels of PD-1 expression observed on Ki67+ CD8+ T cells during CHI were associated with reduced clonal turnover whereas the low levels of PD-1 expression during AHI and as well as in treated and uninfected donors did not affect the capacity of CD8+ T cells to proliferate.

Altogether, our results clearly identify a dual role for PD-1 as both an activation marker during AHI and an exhaustion marker in CD8+ T cells during CHI as measured by the different levels of expression of this receptor in combination with inverse relative levels of Ki67 expression.

Expression of PD-1 is associated with impaired survival and IL-7 responsiveness in CD4+ and CD8+ memory T cells isolated from chronically HIV-1 infected subjects

In order to determine if PD-1 expression identifies cells that show impaired capacity to survive, we measured the baseline expression levels of the survival factor Bcl-2 in ex-vivo total CD4+ and CD8+ memory T cells expressing high and low levels of PD-1 (Fig. 6A). Basal Bcl-2 levels were significantly lower in PD-1high cells when compared to their PD-1low counterparts in the CD4+ (p<0.01 for CHI subjects) as well as in the CD8+ (p<0.05 for CHI subjects and controls) memory compartment (Fig. 6B). This suggested that PD-1 is preferentially expressed by cells endowed with reduced survival capacity, in infected as well as in uninfected individuals.

Figure 6.

Figure 6

Baseline Bcl-2 and IL-7 induced Stat-5 phosphorylation in PD-1high and PD-1low CD45RA memory T cells. A. Representative dot plot analysis showing Bcl-2 expression in PD-1high and PD-1low CD45RA CD4+ or CD8+ T cells. B. Bcl-2 expression in PD-1high and PD-1low CD45RA CD4+ or CD8+ T cells of HIV-1 infected individuals and controls. Each symbol represents one subject: chronic-untreated (n=8; red symbol), and uninfected (n=5; green symbol) subjects. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001. C. Representative dot plot analysis showing pStat5a expression in PD-1high and PD-1low CD45RA CD4+ or CD8+ T cells. D. pStat5a expression in PD-1high and PD-1low CD45RA CD4+ or CD8+ T cells of HIV-1 infected individuals and controls. Each symbol represents one subject: chronic-untreated (n=8; red symbol), and uninfected (n=5; green symbol) subjects. Means are shown as horizontal bars. Statistical significance was determined using the Mann-Whitney U test. * p<0.05, ** p<0.001, *** p<0.0001.

We next evaluated the capacity of memory CD4+ and CD8+ T cells isolated from control individuals and CHI subjects to respond to IL-7 stimulation in vitro by measuring Stat5a phosphorylation upon short-term exposure to the cytokine (Fig. 6C). Memory CD8+ and CD4+ T cells from CHI subjects, irrespective of their level of PD-1 expression, showed lower levels of Stat5a phosphorylation upon IL-7 stimulation when compared to uninfected controls (p<0.05 with the exception of the PD-1high CD4+ memory cells), indicating that HIV infection is associated with impaired T cell responses to IL-7 stimulation. Of note, as this defect is observed in the PD-1high and PD-1low fractions indicates that, in addition to PD-1, other markers may identify cells impaired in their capacity to respond to the homeostatic signal conferred by IL-7 during chronic HIV-1 infection. More importantly, PD-1high memory CD8+ T cells from all study subjects (controls and CHI) displayed lower levels of Stat5a phosphorylation upon IL-7 stimulation when compared to their PD-1low counterparts, indicating that PD-1 identifies memory CD8+ T cells with impaired response to this cytokine. Altogether, our observations indicate that PD-1 expression, which is increased during HIV-1 infection, is associated with a defect in the response to the homeostatic signal conferred by IL-7.

Discussion

We and other groups had previously shown that the up regulation of PD-1 on CD4+ and CD8+ T cells leads to their functional exhaustion during CHI (3034). In the present study we demonstrate that PD-1 is already significantly up regulated on CD4+ and CD8+ T cells during AHI. Moreover, our results indicate that PD-1 is up regulated on all T cell subsets including naïve cells, which have not encountered their cognate antigen yet, as reported in other studies investigating PD-1 expression on T cell subsets during chronic viral infections (4143). A number of antigen-dependent and -independent mechanisms have been proposed for the global up regulation of PD-1 on T cells during chronic HIV-1 infection including residual levels of viral replication (44), inflammatory cytokines (16) and acute homeostatic proliferation of T cells (45). One additional mechanism was proposed by Kinter, et al. who showed that homeostatic cytokines such as IL-2, IL-7, and IL-15 induce the up regulation of PD-1 expression on purified T cells (46). This cytokine-induced PD-1 expression does not impair cytokine-driven events such as proliferation or survival whereas it limits cytokine production after TCR engagement. Moreover, the authors showed that IL-2 administration in successfully treated subjects did not interfere with the ability of the PD-1+ cells to proliferate in vivo (46). However, in chronically infected individuals, the dose and multiplicity of homeostatic cytokines as well as the constant triggering of homeostatic mechanisms may have induced higher level of PD-1 expression on T cells. These higher levels of PD-1 could reach a value that will impair cytokine-driven proliferation as shown by the decrease in Ki67+ T cells.

The size of the peripheral T cell pool is remarkably stable, reflecting precise regulation of cellular proliferation, survival and apoptosis to maintain anatomical niches and the functional diversity needed to respond to pathogenic threats. Homeostasis refers to the capacity of the immune system to preserve its internal equilibrium in T cell subsets number, diversity and distribution, allowing it to return to a normal steady state following perturbation such as infections and lymphopenia. In our study, we observed a breakdown of T cell homeostasis at all stages of the disease. Acutely infected individuals showed an expansion of highly differentiated TEM cells in both the CD4 and the CD8 compartment, which is the signature of most acute viral infections, but also a dramatic diminution in frequencies of CD8+ TN cells and the massive expansion of CD8+ TTM cells. Chronically infected subjects clearly showed a more differentiated profile having lower frequencies of TCM cells and higher frequencies of TEM cells in both the CD4 and CD8 compartment. PD-1 up regulation and continuous PD-L1 engagement during HIV-1 infection could impact peripheral blood T cell subset homeostasis by impairing T cell survival, homeostatic self-renewal, differentiation capacity, de novo priming as well as the functional effector responses to antigen (3034, 47, 48). PD-1 is a sensitive marker of TCR activation as well as tolerance induction following sustained activation by PD-L1/L2, and in this study we find that it represents a marker for the breakdown of naïve/memory T cell subset homeostasis in HIV-1 infection.

Focusing more specifically on the transition from AHI to CHI, our findings provide further insight into potential mechanisms by which immune control in infected individuals might be compromised. Strikingly, HIV-1 chronically infected individuals have an accumulation of naïve cells in both the CD4 and CD8 compartment when compared to acutely infected individuals. As it has been well documented that chronically infected individuals have defective thymic function, this suggests that the accumulation of naïve cells is due to impaired priming and not to the over production of recent thymic emigrants cells from the thymus (22, 49). This could have a huge impact on the diversity of the T cell repertoire of chronically infected individuals, which may result in a less polyclonal repertoire than primary infected individuals. This is consistent with the observation that chronically infected individuals also display much more viral diversity than acutely infected individuals (50, 51). A reduction in the diversity of antigen-specific repertoire might contribute to an inability to control viral infection in chronically infected subjects. Notably, PD-1 up regulation on naïve T cell might contribute to progressive immunodeficiency in HIV-1 disease and might limit the effectiveness of therapeutic vaccine strategies in HIV-1 infection by preventing the activation and expansion of protective T cell responses. Furthermore, chronically infected individuals also have lower numbers of TTM cells than acutely infected individuals. Our finding that both the CD4 and CD8 TTM populations express the highest levels of PD-1 is significant since TTM cells constitute a pool of highly proliferative and polyfunctional cells capable of generating high numbers of effector cells and therefore capable of boosting and sustaining the immune response. Importantly, TTM cells of chronically infected individuals express much higher levels of PD-1 than acutely infected individuals, suggesting that those PD-1high cells may be more susceptible to PD-1 mediated apoptosis (32) or may be impaired in their proliferative capacity. Thus, the accumulation of TN cells and diminished frequency of TTM cells might explain the lack of self-renewal and non-functional anti-viral CD8+ T cells associated with the loss of virus control during CHI compared to AHI.

We next assessed in vivo proliferation of T cell subsets by measuring Ki67 expression during HIV-1 infection. In the CD4 compartment, the percentage of Ki67+ cells in all naïve/memory subpopulations is similar in early and late HIV-1 infection. In the CD8 compartment, there are much higher frequencies of Ki67+ proliferating cells in AHI than in CHI; this being true for total CD8+ T cells but also for each naïve/memory T cell subsets. We observed that levels of PD-1 were the same on CD8+ Ki67+ T cells and CD8+ Ki67 T cells in uninfected and primary infected individuals. In contrast, in chronically infected individuals PD-1 expression was higher on CD8+ Ki67+ T cells than CD8+ Ki67 T cells. The increased levels of PD-1 found on CD8+ Ki67+ T cells in chronically infected individuals could be associated with decreased in vivo proliferative capacity and may partly explain the diminution in percentage of Ki67+ cells in CHI compared to AHI. Of note, our study did not allow us to distinguish between homeostatic and antigen-driven proliferative capacity of Ki67+ cells. Nevertheless, in vitro IL-7 stimulation of PD-1high memory CD8+ T cells showed that these cells have impaired responsiveness to homeostatic cytokine in chronically infected subjects. This suggests that PD-1 expression identifies cells impaired in their capacity to respond to homeostatic signals, in addition to their well-documented defect to respond to antigenic stimulation (3034). In conclusion, PD-1 up regulation on proliferating CD8+ T cells may contribute to their loss of function by altering their proliferative capacity and further limit their ability to expand in sufficient magnitude to exert any control on virus replication. Therefore PD-1 constitutes a marker for the breakdown of T cell distribution in HIV-1 infection.

Our results also show that in addition to the abnormal distribution and loss of T cell function, CD4+ and CD8+ T cells in HIV-1 infected subjects undergo progressive differentiation towards late stages (i.e. CD57+ cells and CD28 cells). In these individuals both CD4+ and CD8+ T cells express CD57 and lose CD28 expression before they have reached late differentiation stages. This abnormal senescence of all naïve/memory subsets clearly contributes to functional immune deficiency. Interestingly, we could not find any significant correlation between PD-1 up regulation and loss of CD28 or CD57 over expression in any of the study groups. Thus, during HIV-1 infection, the accumulation of highly differentiated cells within subsets that usually encompass non-differentiated cells did not correlate with PD-1 up regulation and could partly explain the inability to control viremia.

Recurrent differences were observed between the CD4+ and CD8+ T cell compartments at all stages of the disease. Those include cellular subsets distribution, differentiation/senescence status, loss of function and/or reconstitution after HAART treatment. In the CD4 compartment, we observed barely any differences between acute and chronic infections, suggesting that functional impairment reaches a plateau within 1–3 months after infection. Importantly, our data show that initiation of HAART results in the normalization of almost all parameters in the CD4 compartment of virally suppressed subjects who showed restoration of their CD4+ T cells counts (>400 cells/μl). Of note, the down regulation of PD-1 expression in HAART treated individuals to expression levels seen in uninfected controls has been proposed as a sensitive marker of immune reconstitution in successfully treated subjects (52). Regarding the CD8 compartment, major differences are observed between acute and chronic infections, the most obvious one being the loss of proliferative capacity. Moreover, HAART only partially restores the CD8 compartment, suggesting that longer treatment duration may be needed to reach normalization. This could be explained by the fact that the CD8 compartment is not depleted but is highly impaired in function. In addition, persistent low levels of viral replication in anatomical reservoirs may sustain CD8+ T cell activation and dysfunction (44).

In conclusion, this study provides a comprehensive characterization of phenotypic changes in T cell subsets during HIV-1 infection that reflect different mechanisms involved in the loss of CD4+ and CD8+ T cell function during the acute and chronic stages of HIV-1 infection. Specifically, changes in CD8+ T cell subsets profiles that begin in acute infection and that become fixed in chronic infection, even after initiation of HAART, reveal the profound loss of functional effector cells and the proliferative capability of naïve and memory subsets that could contribute to the eventual failure to control virus replication. Future work to identify the antigen-specificities of CD8+ PD1high TTM cells that are functionally exhausted and the role of PD1-PD-L1 signaling for the establishment of latency in CD4+ T cells will be crucial for the design of therapeutic strategies aimed at blocking PD-1 signaling and other co-inhibitory molecules (53). Our findings here strongly suggest that therapeutic interventions should be initiated early in acute infection before homeostatic mechanisms become deregulated and immune paralysis ensues.

Supplementary Material

1
2
3
4
5

Acknowledgments

The authors thank Colleen Cooper for critical review of this manuscript and Maryse Lainesse for technical assistance.

Grant support: This work was supported by grants awarded to R.-P.S. from the CIHR and the National Institute of Health. J.-P.R. is a clinician-scientist supported by Fonds de Recherche en Santé du Québec. Rémi Fromentin is supported by the American Foundation for AIDS Research (amfAR, fellowship number 108264).

Abbreviations used in this paper

AHI

acute HIV-1 infection

CHI

chronic HIV-1 infection

HAART

highly active antiretroviral therapy

References

  • 1.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. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 2.Giorgi JV, Liu Z, Hultin LE, Cumberland WG, Hennessey K, Detels R. Elevated levels of CD38+ CD8+ T cells in HIV infection add to the prognostic value of low CD4+ T cell levels: results of 6 years of follow-up. The Los Angeles Center, Multicenter AIDS Cohort Study. J Acquir Immune Defic Syndr. 1993;6:904–912. [PubMed] [Google Scholar]
  • 3.Liu Z, Cumberland WG, Hultin LE, Prince HE, Detels R, Giorgi JV. Elevated CD38 antigen expression on CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count, soluble immune activation markers, or combinations of HLA-DR and CD38 expression. J Acquir Immune Defic Syndr Hum Retrovirol. 1997;16:83–92. doi: 10.1097/00042560-199710010-00003. [DOI] [PubMed] [Google Scholar]
  • 4.Cohen Stuart JW, Hazebergh MD, Hamann D, Otto SA, Borleffs JC, Miedema F, Boucher CA, de Boer RJ. The dominant source of CD4+ and CD8+ T-cell activation in HIV infection is antigenic stimulation. J Acquir Immune Defic Syndr. 2000;25:203–211. doi: 10.1097/00126334-200011010-00001. [DOI] [PubMed] [Google Scholar]
  • 5.Hazenberg MD, Stuart JW, Otto SA, Borleffs JC, Boucher CA, de Boer RJ, Miedema F, Hamann D. T-cell division in human immunodeficiency virus (HIV)-1 infection is mainly due to immune activation: a longitudinal analysis in patients before and during highly active antiretroviral therapy (HAART) Blood. 2000;95:249–255. [PubMed] [Google Scholar]
  • 6.Sousa AE, Carneiro J, Meier-Schellersheim M, Grossman Z, Victorino RM. CD4 T cell depletion is linked directly to immune activation in the pathogenesis of HIV-1 and HIV-2 but only indirectly to the viral load. J Immunol. 2002;169:3400–3406. doi: 10.4049/jimmunol.169.6.3400. [DOI] [PubMed] [Google Scholar]
  • 7.Gougeon ML. To kill or be killed: how HIV exhausts the immune system. Cell Death Differ. 2005;12(Suppl 1):845–854. doi: 10.1038/sj.cdd.4401616. [DOI] [PubMed] [Google Scholar]
  • 8.Kedzierska K, Crowe SM. Cytokines and HIV-1: interactions and clinical implications. Antivir Chem Chemother. 2001;12:133–150. doi: 10.1177/095632020101200301. [DOI] [PubMed] [Google Scholar]
  • 9.Papagno L, Spina CA, Marchant A, Salio M, Rufer N, Little S, Dong T, Chesney G, Waters A, Easterbrook P, Dunbar PR, Shepherd D, Cerundolo V, Emery V, Griffiths P, Conlon C, McMichael AJ, Richman DD, Rowland-Jones SL, Appay V. Immune activation and CD8+ T-cell differentiation towards senescence in HIV-1 infection. PLoS Biol. 2004;2:E20. doi: 10.1371/journal.pbio.0020020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Palmer BE, Blyveis N, Fontenot AP, Wilson CC. Functional and phenotypic characterization of CD57+CD4+ T cells and their association with HIV-1-induced T cell dysfunction. J Immunol. 2005;175:8415–8423. doi: 10.4049/jimmunol.175.12.8415. [DOI] [PubMed] [Google Scholar]
  • 11.Appay V, Zaunders JJ, Papagno L, Sutton J, Jaramillo A, Waters A, Easterbrook P, Grey P, Smith D, McMichael AJ, Cooper DA, Rowland-Jones SL, Kelleher AD. Characterization of CD4(+) CTLs ex vivo. J Immunol. 2002;168:5954–5958. doi: 10.4049/jimmunol.168.11.5954. [DOI] [PubMed] [Google Scholar]
  • 12.Giorgi JV, Hultin LE, McKeating JA, Johnson TD, Owens B, Jacobson LP, Shih R, Lewis J, Wiley DJ, Phair JP, Wolinsky SM, Detels R. Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis. 1999;179:859–870. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
  • 13.Hazenberg MD, Otto SA, van Benthem BH, Roos MT, Coutinho RA, Lange JM, Hamann D, Prins M, Miedema F. Persistent immune activation in HIV-1 infection is associated with progression to AIDS. Aids. 2003;17:1881–1888. doi: 10.1097/00002030-200309050-00006. [DOI] [PubMed] [Google Scholar]
  • 14.Koning FA, Otto SA, Hazenberg MD, Dekker L, Prins M, Miedema F, Schuitemaker H. Low-level CD4+ T cell activation is associated with low susceptibility to HIV-1 infection. J Immunol. 2005;175:6117–6122. doi: 10.4049/jimmunol.175.9.6117. [DOI] [PubMed] [Google Scholar]
  • 15.Zeng M, Smith AJ, Wietgrefe SW, Southern PJ, Schacker TW, Reilly CS, Estes JD, Burton GF, Silvestri G, Lifson JD, Carlis JV, Haase AT. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J Clin Invest. 2011;121:998–1008. doi: 10.1172/JCI45157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sedaghat AR, German J, Teslovich TM, Cofrancesco J, Jr, Jie CC, Talbot CC, Jr, Siliciano RF. Chronic CD4+ T-cell activation and depletion in human immunodeficiency virus type 1 infection: type I interferon-mediated disruption of T-cell dynamics. J Virol. 2008;82:1870–1883. doi: 10.1128/JVI.02228-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Smith AJ, Li Q, Wietgrefe SW, Schacker TW, Reilly CS, Haase AT. Host genes associated with HIV-1 replication in lymphatic tissue. J Immunol. 2010;185:5417–5424. doi: 10.4049/jimmunol.1002197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Douek DC, Picker LJ, Koup RA. T cell dynamics in HIV-1 infection. Annu Rev Immunol. 2003;21:265–304. doi: 10.1146/annurev.immunol.21.120601.141053. [DOI] [PubMed] [Google Scholar]
  • 19.Douek DC. Disrupting T-cell homeostasis: how HIV-1 infection causes disease. AIDS Rev. 2003;5:172–177. [PubMed] [Google Scholar]
  • 20.Munier ML, Kelleher AD. Acutely dysregulated, chronically disabled by the enemy within: T-cell responses to HIV-1 infection. Immunol Cell Biol. 2007;85:6–15. doi: 10.1038/sj.icb.7100015. [DOI] [PubMed] [Google Scholar]
  • 21.Dion ML, Poulin JF, Bordi R, Sylvestre M, Corsini R, Kettaf N, Dalloul A, Boulassel MR, Debre P, Routy JP, Grossman Z, Sekaly RP, Cheynier R. HIV infection rapidly induces and maintains a substantial suppression of thymocyte proliferation. Immunity. 2004;21:757–768. doi: 10.1016/j.immuni.2004.10.013. [DOI] [PubMed] [Google Scholar]
  • 22.Douek DC, McFarland RD, Keiser PH, Gage EA, Massey JM, Haynes BF, Polis MA, Haase AT, Feinberg MB, Sullivan JL, Jamieson BD, Zack JA, Picker LJ, Koup RA. Changes in thymic function with age and during the treatment of HIV infection. Nature. 1998;396:690–695. doi: 10.1038/25374. [DOI] [PubMed] [Google Scholar]
  • 23.Schacker TW, Nguyen PL, Beilman GJ, Wolinsky S, Larson M, Reilly C, Haase AT. Collagen deposition in HIV-1 infected lymphatic tissues and T cell homeostasis. J Clin Invest. 2002;110:1133–1139. doi: 10.1172/JCI16413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Okoye A, Meier-Schellersheim M, Brenchley JM, Hagen SI, Walker JM, Rohankhedkar M, Lum R, Edgar JB, Planer SL, Legasse A, Sylwester AW, Piatak M, Jr, Lifson JD, Maino VC, Sodora DL, Douek DC, Axthelm MK, Grossman Z, Picker LJ. Progressive CD4+ central memory T cell decline results in CD4+ effector memory insufficiency and overt disease in chronic SIV infection. J Exp Med. 2007;204:2171–2185. doi: 10.1084/jem.20070567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rethi B, Fluur C, Atlas A, Krzyzowska M, Mowafi F, Grutzmeier S, De Milito A, Bellocco R, Falk KI, Rajnavolgyi E, Chiodi F. Loss of IL-7Ralpha is associated with CD4 T-cell depletion, high interleukin-7 levels and CD28 down-regulation in HIV infected patients. Aids. 2005;19:2077–2086. doi: 10.1097/01.aids.0000189848.75699.0f. [DOI] [PubMed] [Google Scholar]
  • 26.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
  • 27.Pentcheva-Hoang T, Chen L, Pardoll DM, Allison JP. Programmed death-1 concentration at the immunological synapse is determined by ligand affinity and availability. Proc Natl Acad Sci USA. 2007;104:17765–17770. doi: 10.1073/pnas.0708767104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Carter L, Fouser LA, Jussif J, Fitz L, Deng B, Wood CR, Collins M, Honjo T, Freeman GJ, Carreno BM. PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2. Eur J Immunol. 2002;32:634–643. doi: 10.1002/1521-4141(200203)32:3<634::AID-IMMU634>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 30.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. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–1202. doi: 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
  • 31.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. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–354. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
  • 32.Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, Precopio ML, Schacker T, Roederer M, Douek DC, Koup RA. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J Exp Med. 2006;203:2281–2292. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.D’Souza M, Fontenot AP, Mack DG, Lozupone C, Dillon S, Meditz A, Wilson CC, Connick E, Palmer BE. Programmed death 1 expression on HIV-specific CD4+ T cells is driven by viral replication and associated with T cell dysfunction. J Immunol. 2007;179:1979–1987. doi: 10.4049/jimmunol.179.3.1979. [DOI] [PubMed] [Google Scholar]
  • 34.Zhang JY, Zhang Z, Wang X, Fu JL, Yao J, Jiao Y, Chen L, Zhang H, Wei J, Jin L, Shi M, Gao GF, Wu H, Wang FS. PD-1 up-regulation is correlated with HIV-specific memory CD8+ T-cell exhaustion in typical progressors but not in long-term nonprogressors. Blood. 2007;109:4671–4678. doi: 10.1182/blood-2006-09-044826. [DOI] [PubMed] [Google Scholar]
  • 35.Velu V, Titanji K, Zhu B, Husain S, Pladevega A, Lai L, Vanderford TH, Chennareddi L, Silvestri G, Freeman GJ, Ahmed R, Amara RR. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009;458:206–210. doi: 10.1038/nature07662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Younes SA, Trautmann L, Yassine-Diab B, Kalfayan LH, Kernaleguen AE, Cameron TO, Boulassel R, Stern LJ, Routy JP, Grossman Z, Dumont AR, Sekaly RP. The duration of exposure to HIV modulates the breadth and the magnitude of HIV-specific memory CD4+ T cells. J Immunol. 2007;178:788–797. doi: 10.4049/jimmunol.178.2.788. [DOI] [PubMed] [Google Scholar]
  • 37.Miller JD, Masopust D, Wherry EJ, Kaech S, Silvestri G, Ahmed R. Differentiation of CD8 T cells in response to acute and chronic viral infections: implications for HIV vaccine development. Curr Drug Targets Infect Disord. 2005;5:121–129. doi: 10.2174/1568005054201544. [DOI] [PubMed] [Google Scholar]
  • 38.Gamberg J, Pardoe I, Bowmer MI, Howley C, Grant M. Lack of CD28 expression on HIV-specific cytotoxic T lymphocytes is associated with disease progression. Immunol Cell Biol. 2004;82:38–46. doi: 10.1111/j.1440-1711.2004.01204.x. [DOI] [PubMed] [Google Scholar]
  • 39.Appay V, Dunbar PR, Callan M, Klenerman P, Gillespie GM, Papagno L, Ogg GS, King A, Lechner F, Spina CA, Little S, Havlir DV, Richman DD, Gruener N, Pape G, Waters A, Easterbrook P, Salio M, Cerundolo V, McMichael AJ, Rowland-Jones SL. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med. 2002;8:379–385. doi: 10.1038/nm0402-379. [DOI] [PubMed] [Google Scholar]
  • 40.Brenchley JM, Karandikar NJ, Betts MR, Ambrozak DR, Hill BJ, Crotty LE, Casazza JP, Kuruppu J, Migueles SA, Connors M, Roederer M, Douek DC, Koup RA. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood. 2003;101:2711–2720. doi: 10.1182/blood-2002-07-2103. [DOI] [PubMed] [Google Scholar]
  • 41.Shen T, Zheng J, Xu C, Liu J, Zhang W, Lu F, Zhuang H. PD-1 expression on peripheral CD8+ TEM/TEMRA subsets closely correlated with HCV viral load in chronic hepatitis C patients. Virol J. 2010;7:310. doi: 10.1186/1743-422X-7-310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Shen T, Zheng J, Liang H, Xu C, Chen X, Zhang T, Xu Q, Lu F. Characteristics and PD-1 expression of peripheral CD4+CD127loCD25hiFoxP3+ Treg cells in chronic HCV infected-patients. Virol J. 2011;8:279. doi: 10.1186/1743-422X-8-279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rosignoli G, Lim CH, Bower M, Gotch F, Imami N. Programmed death (PD)-1 molecule and its ligand PD-L1 distribution among memory CD4 and CD8 T cell subsets in human immunodeficiency virus-1-infected individuals. Clin Exp Immunol. 2009;157:90–97. doi: 10.1111/j.1365-2249.2009.03960.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Salisch NC, Kaufmann DE, Awad AS, Reeves RK, Tighe DP, Li Y, Piatak M, Jr, Lifson JD, Evans DT, Pereyra F, Freeman GJ, Johnson RP. Inhibitory TCR coreceptor PD-1 is a sensitive indicator of low-level replication of SIV and HIV-1. J Immunol. 2010;184:476–487. doi: 10.4049/jimmunol.0902781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lin SJ, Peacock CD, Bahl K, Welsh RM. Programmed death-1 (PD-1) defines a transient and dysfunctional oligoclonal T cell population in acute homeostatic proliferation. J Exp Med. 2007;204:2321–2333. doi: 10.1084/jem.20062150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, O’Shea MA, Fauci AS. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol. 2008;181:6738–6746. doi: 10.4049/jimmunol.181.10.6738. [DOI] [PubMed] [Google Scholar]
  • 47.Shankar EM, Che KF, Messmer D, Lifson JD, Larsson M. Expression of a broad array of negative costimulatory molecules and Blimp-1 in T cells following priming by HIV-1 pulsed dendritic cells. Mol Med. 2011;17:229–240. doi: 10.2119/molmed.2010.00175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 49.Douek DC, Betts MR, Hill BJ, Little SJ, Lempicki R, Metcalf JA, Casazza J, Yoder C, Adelsberger JW, Stevens RA, Baseler MW, Keiser P, Richman DD, Davey RT, Koup RA. Evidence for increased T cell turnover and decreased thymic output in HIV infection. J Immunol. 2001;167:6663–6668. doi: 10.4049/jimmunol.167.11.6663. [DOI] [PubMed] [Google Scholar]
  • 50.Abrahams MR, Anderson JA, Giorgi EE, Seoighe C, Mlisana K, Ping LH, Athreya GS, Treurnicht FK, Keele BF, Wood N, Salazar-Gonzalez JF, Bhattacharya T, Chu H, Hoffman I, Galvin S, Mapanje C, Kazembe P, Thebus R, Fiscus S, Hide W, Cohen MS, Karim SA, Haynes BF, Shaw GM, Hahn BH, Korber BT, Swanstrom R, Williamson C. Quantitating the multiplicity of infection with human immunodeficiency virus type 1 subtype C reveals a non-poisson distribution of transmitted variants. J Virol. 2009;83:3556–3567. doi: 10.1128/JVI.02132-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fischer W, V, Ganusov V, Giorgi EE, Hraber PT, Keele BF, Leitner T, Han CS, Gleasner CD, Green L, Lo CC, Nag A, Wallstrom TC, Wang S, McMichael AJ, Haynes BF, Hahn BH, Perelson AS, Borrow P, Shaw GM, Bhattacharya T, Korber BT. Transmission of single HIV-1 genomes and dynamics of early immune escape revealed by ultra-deep sequencing. PLoS One. 2010;5:e12303. doi: 10.1371/journal.pone.0012303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Grabmeier-Pfistershammer K, Steinberger P, Rieger A, Leitner J, Kohrgruber N. Identification of PD-1 as a unique marker for failing immune reconstitution in HIV-1-infected patients on treatment. J Acquir Immune Defic Syndr. 2011;56:118–124. doi: 10.1097/QAI.0b013e3181fbab9f. [DOI] [PubMed] [Google Scholar]
  • 53.Kline J, Gajewski TF. Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer. Curr Opin Investig Drugs. 2010;11:1354–1359. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3
4
5

RESOURCES