Immune Netw. 2010 Aug;10(4):120-125. English.
Published online Aug 31, 2010.
Copyright © 2010 The Korean Association of Immunologists
Review

T-Cell Dysfunction and Inhibitory Receptors in Hepatitis C Virus Infection

Jino Lee, William I. Suh and Eui-Cheol Shin
    • Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Korea.
Received August 18, 2010; Accepted August 24, 2010.

This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Dysfunction of the virus-specific T cells is a cardinal feature in chronic persistent viral infections such as one caused by hepatitis C virus (HCV). In chronic HCV infection, virus-specific dysfunctional CD8 T cells often overexpress various inhibitory receptors. Programmed cell death 1 (PD-1) was the first among these inhibitory receptors that were identified to be overexpressed in functionally impaired T cells. The roles of other inhibitory receptors such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and T cell immunoglobulin and mucin domain-containing molecule 3 (Tim-3) have also been demonstrated in T-cell dysfunctions that occur in chronic HCV patients. Blocking these inhibitory receptors in vitro restores the functions of HCV-specific CD8 T cells and allows enhanced proliferation, cytolytic activity and cytokine production. Therefore, the blockade of the inhibitory receptors is considered as a novel strategy for the treatment of chronic HCV infection.

Keywords
Hepatitis C virus; CD8 T cells; T-cell dysfunction; PD-1; CTLA-4; Tim-3

INTRODUCTION

Hepatitis C virus (HCV) is a positive-sense single-stranded RNA virus of the genus Hepacivirus in the family Flaviviridae, and it infects 170 million people worldwide (1). About 10~60% of the patients clear HCV spontaneously during the acute phase of the infection (2), while the others develop chronic persistent HCV infection that eventually leads to liver cirrhosis and hepatocellular carcinoma (3). Spontaneous resolution of HCV infection correlates with robust and sustained responses of the virus-specific T cells as demonstrated in humans (4-6) and in chimpanzees (7, 8), the sole animal model of HCV infection. On the other hand, the progression towards chronic HCV infection is associated with weak and transient responses of the virus-specific T cells (4-8). Various dysfunctions of the HCV-specific T cells, such as inefficient proliferation, cytolytic activity, and cytokine production, are commonly observed during the chronic stage of HCV infection (reviewed in 9, 10). Impaired cellular immune responses have been attributed to the mutations within the T-cell epitopes (11-13), a deviated differentiation of T cells (14) and suppressive functions of the regulatory T cells (15). Dysfunctional T cells are also observed in other chronic persistent viral infections such as hepatitis B virus (HBV), human immunodeficiency virus (HIV) in humans, and lymphocytic choriomeningitis virus (LCMV) infection in mice (16).

A novel mechanism of T-cell dysfunction was recently demonstrated in a murine model of chronic LCMV infection (17). It was found that the expression of programmed cell death 1 (PD-1) was up-regulated on dysfunctional LCMV-specific CD8 T cells in mice (17). In vivo blockade of the interaction between PD-1 and its ligand, PD-L1, restored the functions of LCMV-specific CD8 T cells and reduced the viral titer (17). This influential discovery led to extensive investigations of the role of PD-1 in the regulation of T cells in human chronic viral infections (16). More recently, other inhibitory receptors such as cytotoxic T lymphocyte asoociated antigen 4 (CTLA-4) and T cell immunoglobulin and mucin domain containing molecule 3 (Tim-3) have also been studied as the factors that can cause T-cell impairments in chronic viral infections. In this review, the roles of various inhibitory receptors in T-cell dysfunction found in chronic HCV infection are summarized.

THE ROLE OF PD-1 IN HCV INFECTION

PD-1 is one of the inhibitory receptors which are expressed on the T cells. It has two known ligands, PD-L1 and PD-L2, which are members of B7 family. Upon binding to its ligands, PD-1 confers inhibitory signal to the T cells by recruiting SH2-containing phosphatases, SHP-1 and SHP-2, to its immunoreceptor tyrosine-based switch motif (ITSM). Recruited phospatases then block the T-cell receptor (TCR)-mediated activatory signal at proximal site (16).

The role of PD-1 in virus-specific T cells in chronic viral infections was first identified in a murine model of chronic LCMV infection (17). As in chronic LCMV infection, the expression of PD-1 is similarly upregulated on the virus-specific CD8 T cells in chronic HCV infection, and HCV-specific PD-1high T cells are functionally impaired (18-20). In addition, a blockade of PD-1/PD-L1 interaction restores T-cell functions such as proliferation, cytolytic activity and cytokine (IFN-γ and TNF-α) production (18-20). The PD-1high dysfunctional CD8 T cells express low levels of CD127, a marker of memory precursors, and high levels of CD57, a T-cell senescence marker (18, 20). PD-1highCD127low HCV-specific CD8 T cells are known to frequently undergo apoptosis (21). PD-1 expression is likely to be influenced by the location of HCV-specific CD8 T cells in vivo, since HCV-specific CD8 T cells in the liver have a tendency to express higher levels of PD-1 than those found in the peripheral blood (22). In addition, PD-1/PD-L1 blockade was able to functionally restore HCV-specific CD8 T cells originating from the peripheral blood, but not those found in the liver (22). The dissimilarities among the virus-specific CD8 T cells found in different in vivo compartments need to be considered in further studies.

The role of PD-1 was also studied in the acute stage of HCV infection. Specifically, the relationship between the PD-1 expression and the outcome of the acute HCV infection was questioned. Recent studies showed that the progression of acute HCV infection to the chronic stage is associated with a high level of PD-1 on HCV-specific CD8 T cells during the acute infection, and the clearance of HCV infection is associated with lower levels of PD-1 expression (23, 24). However, at least one other study reported that the high level of PD-1 during the acute HCV infection is irrespective of the outcome of HCV infection (25). In a chimpanzee model of acute HCV infection, intrahepatic levels of PD-1 were determined by prospective liver biopsy and real-time PCR, and high mRNA levels of PD-1 were found to be associated with the development of chronic HCV infection (26). Intriguingly, the PD-1 levels in HCV-specific CD8 T cells declined through the escaping mutation of cognate T-cell epitopes even in the chronic stage of HCV infection (23). This implies that the high PD-1 levels on virus-specific CD8 T cells are maintained by persistent TCR stimulation, which henceforth explains why high PD-1 expression is routinely observed in chronic persistent viral infections.

Very recently, PD-1 expression was studied in the experimental vaccine trials and subsequent HCV challenge in chimpanzees (Shin et al., unpublished data). In this study, the phenotypes of HCV-specific CD8 T cells were analyzed in HCV-challenged chimpanzees, which were part of a previously published adenovirus/DNA-based HCV NS3-NS5 vaccine study (27). HCV-specific CD8 T cells from the vaccinated chimpanzees displayed lower levels of PD-1 and a greater ability to secrete IFN-γ than those from the control group. Consistent with these findings, intrahepatic mRNA levels of PD-1 and PD-L1 were significantly lower in the vaccinated chimpanzees than in the control chimpanzees. These data showed that the low expressions of PD-1 and PD-L1 are characteristic features of vaccine-induced resolution of acute HCV infection, and that the attenuation of the PD-1/PD-L1 inhibitory pathway during vaccine-induced HCV clearance may enable HCV-specific CD8 T cells to have enhanced anti-viral functions.

THE ROLE OF CTLA-4 IN HCV INFECTION

CTLA-4 is structurally homologous to CD28, an important T-cell costimulatory molecule, and its expression is upregulated on the activated T cells (28). CTLA-4 exerts T-cell inhibitory functions through diverse mechanisms. CTLA-4 binds to CD80 and CD86, thus competitively inhibiting the interaction between CD28 and B7 molecules. In addition, CTLA-4 recruits phosphatases such as SHP-2 and blocks the signal activated by TCR ligation (28).

Although the blockade of CTLA-4 did not result in a restoration of the T cell functions in chronic LCMV infection in the previous study (17), recent findings on CTLA-4's role in chronic HCV infection showed promising results (22, 29). The HCV-specific CD8 T cells found in the livers of chronic HCV patients did not only overexpress PD-1, but also CTLA-4. Co-expression of PD-1 and CTLA-4 was observed in liver-infiltrating lymphocytes, but not in peripheral blood lymphocytes (29), suggesting the phenotypic differences of virus-specific CD8 T cells in different in vivo compartments. PD-1+CTLA-4+ HCV-specific T cells were profoundly dysfunctional (22). The functions of PD-1+CTLA-4+ HCV-specific CD8 T cells could be restored by a combined blockade of PD-1 and CTLA-4, but not by PD-1 blockade or CTLA-4 blockade alone (29). For the development of a novel therapeutic strategy to restore the functions of HCV-specific CD8 T cells, a combined blockade of multiple inhibitory receptors needs to be done in order to maximize the anti-viral functions of HCV-specific CD8 T cells.

THE ROLE OF Tim-3 IN HCV INFECTION

Tim-3 was originally discovered as a specific marker of Th1 CD4 T cells (30). It has been known that interaction of Tim-3 with its ligand, galectin-9, promotes the cell death of Th1 cells and terminates Th1 responses.

The role of Tim-3 in chronic viral infections was first identified in HIV infection (31). In HIV-infected patients, the frequency of Tim-3+ CD8 T cells increased, and the Tim-3 levels on the T cells correlated positively with the viral titer and inversely with CD4 T cell count (31). The HIV-specific CD8 T cells that overexpress Tim 3 were found to be functionally impaired, and a blockade of Tim-3 restored the functions of HIV-specific CD8 T cells (31).

After this finding, the role of Tim-3 has also been studied in chronic HCV infection (32). Tim-3 is over-expressed on HCV-specific dysfunctional CD8 T cells, and Tim-3+ CD8 T cells are of CD127lowCD57high, phenotype which is identical to that of PD-1+ CD8 T cells in chronic HCV infection (18, 20). Tim-3+PD-1+ HCV-specific CD8 T cells were preferentially enriched in the intrahepatic compartment over the peripheral blood. Importantly, a blockade of Tim-3 resulted in a functional restoration of HCV-specific CD8 T cells, evidenced by increased proliferation and IFN-γ production (32). The role of Tim-3 was also studied in HCV/HIV co-infection (33). Compared to HCV infection alone, the frequency of Tim-3+PD-1+ HCV-specific CD8 T cells was higher in HCV/HIV co-infection, and Tim-3/PD-1 co-expression correlated with liver damage (33). Either a Tim-3 blockade or a PD-1 blockade alone was found to be sufficient in restoring the functions of Tim-3+PD-1+ HCV-specific CD8 T cells. Interestingly, Tim-3+PD-1+ phenotype was more frequent in HCV-specific CD8 T cells than in HIV-specific CD8 T cells, implying the varying degrees of impairments in different virus specific T cells.

THE ROLE OF OTHER INHIBITORY RECEPTORS IN HCV INFECTION

In order to discover the other possible molecules that can potentially downregulate T-cell functions in chronic viral infections, microarray and gene expression profiling were performed in a murine model of LCMV infection (34). Several candidate molecules have been identified, including PD-1, lymphocyte activation gene-3 (LAG-3), 2B4, CD160, CTLA-4, paired immunoglobulin-like receptor B (PIR-B) and GP49B (34). A subsequent study demonstrated the complicated expression patterns of the inhibitory molecules and showed that the co-expression of multiple inhibitory molecules is associated with the severity of the infection (35). Very recently, co-expression of PD-1, 2B4, CD160, killer cell lectin-like receptor G1 (KLRG1), LAG-3 and CTLA-4 on CD8 T cells was studied in chronic HCV infection (36). Co-expression of multiple inhibitory receptors was observed on HCV-specific CD8 T cells and was associated with low levels of CD127 (36).

CONCLUSION

Since the discovery of PD-1 as an inhibitory receptor associated with T-cell dysfunction in chronic LCMV infection, the roles of various inhibitory receptors on virus-specific CD8 T cells have been extensively studied in human chronic viral infections such as HCV, HBV and HIV infections. As blocking the inhibitory receptors in vitro restored the functions of virus-specific T cells, the blockade has been considered as a novel strategy for the treatment of chronic viral infections (37). A recent study evaluated the in vivo blocking effects of anti-PD-1 antibody in macaques infected with simian immunodeficiency virus (SIV) (38). The PD-1 blockade was able to enhance the immune responses and resulted in a significant reduction of viral load and prolonged survival of the infected hosts (38).

However, for an in vivo blockade of the inhibitory receptors to be used in an actual therapy, some possible side effects must be considered. One study found that the infection of PD-L1-/- mice with a chronic LCMV strain was lethal due to severe immunopathologic damage (17), implying the importance of PD-1/PD-L1 in the prevention of virus-induced lethal immunopathology. In particular, liver damage is known to be mediated by T-cell responses in HCV infections, and hence T cell-mediated liver damage may be aggravated by blockades of inhibitory receptors, resulting in lethal hepatitis (39).

As mentioned before, some patients recover from acute HCV infection and exhibit low level of inhibitory receptors, while the others enter the chronic stage of HCV infection that results in upregulation of inhibitory receptors and progressive loss of the T-cell functions. Thus, an important question that arises is the mechanism that influences the body's immune system to choose between those two directions of disease outcome during the acute HCV infection. Perhaps it could be based on the patient's genetic backgrounds, environmental influences, or both. In any case, the potential possibility that the blockade of the inhibitory molecules early on during the acute HCV infection may prevent the disease from progressing toward a more problematic chronic stage should be considered.

The restoration of the patients' own anti-viral immune functions has been pursued as a possible therapy for chronic HCV infection, but all efforts have been unsuccessful to date. The recent advancements in the understandings of the roles of the inhibitory receptors in T-cell dysfunction will hopefully aid greatly in the development of a highly effective therapy for HCV infection.

Notes

The authors have no financial conflict of interest.

ACKNOWLEDGEMENTS

This research was supported by the Korea Research Foundation Grant funded by the Korean Government (KRF-2008-313-E00245) and by the Research Program for New Drug Target Discovery through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (2010-0020471).

References

    1. Shepard CW, Finelli L, Alter MJ. Global epidemiology of hepatitis C virus infection. Lancet Infect Dis 2005;5:558–567.
    1. Kamal SM. Acute hepatitis C: a systematic review. Am J Gastroenterol 2008;103:1283–1297.
    1. Alter HJ, Seeff LB. Recovery, persistence, and sequelae in hepatitis C virus infection: a perspective on long-term outcome. Semin Liver Dis 2000;20:17–35.
    1. Diepolder HM, Zachoval R, Hoffmann RM, Wierenga EA, Santantonio T, Jung MC, Eichenlaub D, Pape GR. Possible mechanism involving T-lymphocyte response to non-structural protein 3 in viral clearance in acute hepatitis C virus infection. Lancet 1995;346:1006–1007.
    1. Lechner F, Wong DK, Dunbar PR, Chapman R, Chung RT, Dohrenwend P, Robbins G, Phillips R, Klenerman P, Walker BD. Analysis of successful immune responses in persons infected with hepatitis C virus. J Exp Med 2000;191:1499–1512.
    1. Thimme R, Oldach D, Chang KM, Steiger C, Ray SC, Chisari FV. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J Exp Med 2001;194:1395–1406.
    1. Cooper S, Erickson AL, Adams EJ, Kansopon J, Weiner AJ, Chien DY, Houghton M, Parham P, Walker CM. Analysis of a successful immune response against hepatitis C virus. Immunity 1999;10:439–449.
    1. Thimme R, Bukh J, Spangenberg HC, Wieland S, Pemberton J, Steiger C, Govindarajan S, Purcell RH, Chisari FV. Viral and immunological determinants of hepatitis C virus clearance, persistence, and disease. Proc Natl Acad Sci USA 2002;99:15661–15668.
    1. Rehermann B, Nascimbeni M. Immunology of hepatitis B virus and hepatitis C virus infection. Nat Rev Immunol 2005;5:215–229.
    1. Dustin LB, Rice CM. Flying under the radar: the immunobiology of hepatitis C. Annu Rev Immunol 2007;25:71–99.
    1. Erickson AL, Kimura Y, Igarashi S, Eichelberger J, Houghton M, Sidney J, McKinney D, Sette A, Hughes AL, Walker CM. The outcome of hepatitis C virus infection is predicted by escape mutations in epitopes targeted by cytotoxic T lymphocytes. Immunity 2001;15:883–895.
    1. Chang KM, Rehermann B, McHutchison JG, Pasquinelli C, Southwood S, Sette A, Chisari FV. Immunological significance of cytotoxic T lymphocyte epitope variants in patients chronically infected by the hepatitis C virus. J Clin Invest 1997;100:2376–2385.
    1. Weiner A, Erickson AL, Kansopon J, Crawford K, Muchmore E, Hughes AL, Houghton M, Walker CM. Persistent hepatitis C virus infection in a chimpanzee is associated with emergence of a cytotoxic T lymphocyte escape variant. Proc Natl Acad Sci USA 1995;92:2755–2759.
    1. 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.
    1. Sugimoto K, Ikeda F, Stadanlick J, Nunes FA, Alter HJ, Chang KM. Suppression of HCV-specific T cells without differential hierarchy demonstrated ex vivo in persistent HCV infection. Hepatology 2003;38:1437–1448.
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 2007;8:239–245.
    1. 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.
    1. Radziewicz H, Ibegbu CC, Fernandez ML, Workowski KA, Obideen K, Wehbi M, Hanson HL, Steinberg JP, Masopust D, Wherry EJ, Altman JD, Rouse BT, Freeman GJ, Ahmed R, Grakoui A. Liver-infiltrating lymphocytes in chronic human hepatitis C virus infection display an exhausted phenotype with high levels of PD-1 and low levels of CD127 expression. J Virol 2007;81:2545–2553.
    1. Penna A, Pilli M, Zerbini A, Orlandini A, Mezzadri S, Sacchelli L, Missale G, Ferrari C. Dysfunction and functional restoration of HCV-specific CD8 responses in chronic hepatitis C virus infection. Hepatology 2007;45:588–601.
    1. Golden-Mason L, Palmer B, Klarquist J, Mengshol JA, Castelblanco N, Rosen HR. Upregulation of PD-1 expression on circulating and intrahepatic hepatitis C virus-specific CD8+ T cells associated with reversible immune dysfunction. J Virol 2007;81:9249–9258.
    1. Radziewicz H, Ibegbu CC, Hon H, Osborn MK, Obideen K, Wehbi M, Freeman GJ, Lennox JL, Workowski KA, Hanson HL, Grakoui A. Impaired hepatitis C virus (HCV)-specific effector CD8+ T cells undergo massive apoptosis in the peripheral blood during acute HCV infection and in the liver during the chronic phase of infection. J Virol 2008;82:9808–9822.
    1. Nakamoto N, Kaplan DE, Coleclough J, Li Y, Valiga ME, Kaminski M, Shaked A, Olthoff K, Gostick E, Price DA, Freeman GJ, Wherry EJ, Chang KM. Functional restoration of HCV-specific CD8 T cells by PD-1 blockade is defined by PD-1 expression and compartmentalization. Gastroenterology 2008;134:1927–1937.
    1. Rutebemberwa A, Ray SC, Astemborski J, Levine J, Liu L, Dowd KA, Clute S, Wang C, Korman A, Sette A, Sidney J, Pardoll DM, Cox AL. High-programmed death-1 levels on hepatitis C virus-specific T cells during acute infection are associated with viral persistence and require preservation of cognate antigen during chronic infection. J Immunol 2008;181:8215–8225.
    1. Urbani S, Amadei B, Tola D, Massari M, Schivazappa S, Missale G, Ferrari C. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J Virol 2006;80:11398–11403.
    1. Kasprowicz V, Schulze Zur Wiesch J, Kuntzen T, Nolan BE, Longworth S, Berical A, Blum J, McMahon C, Reyor LL, Elias N, Kwok WW, McGovern BG, Freeman G, Chung RT, Klenerman P, Lewis-Ximenez L, Walker BD, Allen TM, Kim AY, Lauer GM. High level of PD-1 expression on hepatitis C virus (HCV)-specific CD8+ and CD4+ T cells during acute HCV infection, irrespective of clinical outcome. J Virol 2008;82:3154–3160.
    1. Rollier CS, Paranhos-Baccala G, Verschoor EJ, Verstrepen BE, Drexhage JA, Fagrouch Z, Berland JL, Komurian-Pradel F, Duverger B, Himoudi N, Staib C, Meyr M, Whelan M, Whelan JA, Adams VC, Larrea E, Riezu JI, Lasarte JJ, Bartosch B, Cosset FL, Spaan WJ, Diepolder HM, Pape GR, Sutter G, Inchauspe G, Heeney JL. Vaccine-induced early control of hepatitis C virus infection in chimpanzees fails to impact on hepatic PD-1 and chronicity. Hepatology 2007;45:602–613.
    1. Folgori A, Capone S, Ruggeri L, Meola A, Sporeno E, Ercole BB, Pezzanera M, Tafi R, Arcuri M, Fattori E, Lahm A, Luzzago A, Vitelli A, Colloca S, Cortese R, Nicosia A. A T-cell HCV vaccine eliciting effective immunity against heterologous virus challenge in chimpanzees. Nat Med 2006;12:190–197.
    1. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol 2001;19:565–594.
    1. Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, Gostick E, Price DA, Freeman GJ, Wherry EJ, Chang KM. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathog 2009;5:e1000313.
    1. Meyers JH, Sabatos CA, Chakravarti S, Kuchroo VK. The TIM gene family regulates autoimmune and allergic diseases. Trends Mol Med 2005;11:362–369.
    1. Jones RB, Ndhlovu LC, Barbour JD, Sheth PM, Jha AR, Long BR, Wong JC, Satkunarajah M, Schweneker M, Chapman JM, Gyenes G, Vali B, Hyrcza MD, Yue FY, Kovacs C, Sassi A, Loutfy M, Halpenny R, Persad D, Spotts G, Hecht FM, Chun TW, McCune JM, Kaul R, Rini JM, Nixon DF, Ostrowski MA. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med 2008;205:2763–2779.
    1. Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, Castelblanco N, Kuchroo V, Gretch DR, Rosen HR. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol 2009;83:9122–9130.
    1. Vali B, Jones RB, Sakhdari A, Sheth PM, Clayton K, Yue FY, Gyenes G, Wong D, Klein MB, Saeed S, Benko E, Kovacs C, Kaul R, Ostrowski MA. HCV-specific T cells in HCV/HIV co-infection show elevated frequencies of dual Tim-3/PD-1 expression that correlate with liver disease progression. Eur J Immunol 2010;40:2493–2505.
    1. Wherry EJ, Ha SJ, Kaech SM, Haining WN, Sarkar S, Kalia V, Subramaniam S, Blattman JN, Barber DL, Ahmed R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007;27:670–684.
    1. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 2009;10:29–37.
    1. Bengsch B, Seigel B, Ruhl M, Timm J, Kuntz M, Blum HE, Pircher H, Thimme R. Coexpression of PD-1, 2B4, CD160 and KLRG1 on exhausted HCV-specific CD8+ T cells is linked to antigen recognition and T cell differentiation. PLoS Pathog 2010;6:e1000947.
    1. Chang DY, Shin EC. Immune-based therapy for chronic hepatitis C. J Leukoc Biol 2009;86:33–39.
    1. 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.
    1. Shin EC, Rehermann B. Taking the brake off T cells in chronic viral infection. Nat Med 2006;12:276–277.

Metrics
Share
ORCID IDs
PERMALINK