Releasing CD8+ Treg Mediated Suppresssion Enhances Anti-Viral Immune Response

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2280-2280
Author(s):  
Tobias A.W. Holderried ◽  
Hye-Jung Kim ◽  
Philipp A Lang ◽  
Harvey Cantor

Abstract Recent findings have shown that a small subset of IL-15 dependent CD8+ regulatory T cells is essential for maintenance of self- tolerance and prevention of autoimmune disease in mice (Kim et al., Nature 2010). These CD8+ T cells target CD4+ follicular T helper (TFH) cells through recognition of the murine class Ib MHC molecule Qa-1 (HLA-E in man), resulting in perforin-dependent elimination of target cells and diminished antibody production in the steady state and during disease. This analysis was based on generation of Qa-1 knock-in mice (D227K mice) that harbor a single Qa-1 D→K amino acid exchange point mutation at position 227 that abrogates binding of Qa-1/peptide to the CD8/TCR complex. B6.Qa-1 D227K mutant mice develop severe autoimmune disease marked by generation of autoantibodies to multiple tissues, lymphocyte infiltration into non-lymphoid tissues and lethal glomerulonephritis. Qa-1-restricted CD8+ Treg are characterized by the CD44+CD122+Ly49+ phenotype (Kim et al., PNAS 2011). Here, we analyzed the contribution of CD8+ Treg to modulation of the anti-viral immune response. Virus-specific CD8+ cytotoxic T cells are of central importance for successful control of the Lymphocytic Choriomeningitis Virus (LCMV). LCMV clone 13, however, a genetic variant of LCMV Armstrong, persists in the host and chronic antigen exposure leads to exhaustion of CD8+ T cells and continuous tissue inflammation. The contribution of CD8+ Treg in the anti-viral immune response to acute and chronic viral infection remained elusive so far. We found that CD8+ Treg not only control self-tolerance but also diminish the immune response to viral infection. By comparing wild-type and D227K mutant mice after infection with LCMV Armstrong or LCMV clone 13, we observed in both cases reduced effector CD8+ T cell responses. This was true for polyclonal CD44+CD62L– CD8+ T cells as well as LCMV-specific gp33+ effector CD8+ T cells. During acute infection KLRG1+CD127-CD44+CD62L- cells (short-lived effector CD8+ cells) (Joshi et al., Immunity 2007) were particularly diminished as well as effector cytokines in wild-type mice compared to D227K mice. In contrast, increased effector responses in D227K mice resulted in enhanced control of virus and reduced inflammation of tissues. During chronic infection with LCMV, wild-type mice become severely ill and present with a pronounced clinical phenotype. Increased effector CD8+ T cell immune responses in D227K mice resulted in dramatic alleviation of disease. During late stage of chronic infection, D227K mice showed enhanced virus control and reduced tissue pathology compared with wild-type mice. Interestingly, expression of inhibitory receptors such as PD-1, 2B4 and LAG3 were increased in wild-type mice whereas activating receptors such as NKG2D and KLRG1 were increased in D227K mice, resulting in a memory phenotype in D227K mice compared with exhausted CD8+ T cells in wild-type mice. Adoptive transfer experiments revealed that CD8+ Treg directly suppress CD8+ target cells and thereby inhibit induction of a robust anti-viral response. Taken together, we show that Qa-1-restricted CD8+ Treg have a direct inhibitory effect on effector CD8+ T cells during acute and chronic viral infection, resulting in a more violent disease and diminished recovery. These data suggest that depletion or inactivation of CD8+ Treg represents a potentially effective strategy to enhance anti-viral immunity. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2227-2227
Author(s):  
Shannon A. Carty ◽  
Mercy Gohil ◽  
Erietta Stelekati ◽  
Lauren B. Banks ◽  
E. John Wherry ◽  
...  

Abstract DNA methylation is one of the major epigenetic mechanisms that controls cellular differentiation. The ten-eleven translocation (TET) family of methylcytosine dioxygenases mediates active DNA demethylation through the oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and subsequent intermediates. Here we demonstrate that TET2 regulates CD8+ T cell differentiation in vivo following acute and chronic viral infection. At steady-state, mice with a T-cell specific deletion of TET2 have intact thymic and peripheral T cell populations. Following acute viral infection with LCMV-Armstrong, TET2 loss enhances LCMV-specific CD8+ T cell memory differentiation in a cell-intrinsic manner without disrupting antigen-specific cell expansion or cytokine production. However, TET2-deficient memory CD8+ T cells exhibit altered recall responses with blunted re-expansion, retained expression of phenotypic memory markers and restricted re-expression of activation markers. During chronic viral infection with LCMV-clone 13, TET2 controls CD8+ T cell expansion and alters differentiation. Importantly, though mice with T-cell specific loss of TET2 developed similar levels of CD8+ T cell exhaustion as wild-type mice, TET2 loss specifically reduced PD-1 expression suggesting that TET2 may direct DNA demethylation of the PD-1 locus. Together, our data indicate that TET2 is an important regulator of CD8+ T cells following both acute and chronic viral infections and suggest targeting epigenetic regulators have potential for enhancing antiviral immunity. Disclosures No relevant conflicts of interest to declare.


1996 ◽  
Vol 184 (4) ◽  
pp. 1543-1547 ◽  
Author(s):  
S Sad ◽  
D Kägi ◽  
T R Mosmann

During an immune response, effector CD8+ T cells can kill infected cells by the perforin-dependent pathway. In comparison to CD4+ T cells, which are major sources of cytokines, normal CD8+ T cells produced less interleukin 2 and interferon gamma, and proliferated less vigorously after antigenic stimulation. Killing of target cells was a major cause of these reduced responses, since perforin-deficient CD8+ T cells showed substantially increased cytokine synthesis and proliferation. Cytotoxicity by the alternate Fas pathway also resulted in self-limitation of CD8+ T cell cytokine synthesis. This relationship between cytotoxicity and cytokine synthesis may regulate CD8+ T function in different phases of an immune response.


Viruses ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1189
Author(s):  
David G. Brooks ◽  
Antoinette Tishon ◽  
Michael B. A. Oldstone ◽  
Dorian B. McGavern

During chronic viral infections, CD8 T cells rapidly lose antiviral and immune-stimulatory functions in a sustained program termed exhaustion. In addition to this loss of function, CD8 T cells with the highest affinity for viral antigen can be physically deleted. Consequently, treatments designed to restore function to exhausted cells and control chronic viral replication are limited from the onset by the decreased breadth of the antiviral T cell response. Yet, it remains unclear why certain populations of CD8 T cells are deleted while others are preserved in an exhausted state. We report that CD8 T cell deletion during chronic viral infection can be prevented by therapeutically lowering viral replication early after infection. The initial resistance to deletion enabled long-term maintenance of antiviral cytolytic activity of the otherwise deleted high-affinity CD8 T cells. In combination with decreased virus titers, CD4 T cell help and prolonged interactions with costimulatory molecules B7-1/B7-2 were required to prevent CD8 T cell deletion. Thus, therapeutic strategies to decrease early virus replication could enhance virus-specific CD8 T cell diversity and function during chronic infection.


2020 ◽  
Vol 117 (8) ◽  
pp. 4292-4299 ◽  
Author(s):  
Se Jin Im ◽  
Bogumila T Konieczny ◽  
William H. Hudson ◽  
David Masopust ◽  
Rafi Ahmed

The migratory patterns of virus-specific CD8 T cells during chronic viral infection are not well understood. To address this issue, we have done parabiosis experiments during chronic lymphocytic choriomeningitis virus (LCMV) infection of mice. We found that despite the high frequency of virus-specific CD8 T cells in both lymphoid and nonlymphoid tissues there was minimal migration of virus-specific CD8 T cells between the chronically infected conjoined parabiont mice. This was in contrast to parabionts between mice that had undergone an acute LCMV infection where virus-specific CD8 T cells established equilibrium demonstrating circulation of memory T cells generated after viral clearance. We have identified a population of PD-1+ TCF1+CXCR5+Tim-3- stemlike virus-specific CD8 T cells that reside in lymphoid tissues and act as resource cells for maintaining the T cell response during chronic infection. These are the cells that proliferate and give rise to the more terminally differentiated PD-1+ CXCR5-Tim-3+ CD8 T cells. Both the stemlike CD8 T cells and their terminally differentiated progeny showed minimal migration during chronic infection and the few LCMV-specific CD8 T cells that were present in circulation were the recently emerging progeny from the stemlike CD8 T cells. The PD-1+ TCF1+CXCR5+ stemlike CD8 T cells were truly resident in lymphoid tissues and did not circulate in the blood. We propose that this residency in specialized niches within lymphoid tissues is a key aspect of their biology and is essential for maintaining their quiescence and stemlike program under conditions of a chronic viral infection.


2002 ◽  
Vol 195 (6) ◽  
pp. 695-704 ◽  
Author(s):  
Michel Gilliet ◽  
Yong-Jun Liu

Although CD8 T cell–mediated immunosuppression has been a well-known phenomenon during the last three decades, the nature of primary CD8 T suppressor cells and the mechanism underlying their generation remain enigmatic. We demonstrated that naive CD8 T cells primed with allogeneic CD40 ligand–activated plasmacytoid dendritic cells (DC)2 differentiated into CD8 T cells that displayed poor secondary proliferative and cytolytic responses. By contrast, naive CD8 T cells primed with allogeneic CD40 ligand–activated monocyte-derived DCs (DC1) differentiated into CD8 T cells, which proliferated to secondary stimulation and killed allogeneic target cells. Unlike DC1-primed CD8 T cells that produced large amounts of interferon (IFN)-γ upon restimulation, DC2-primed CD8 T cells produced significant amounts of interleukin (IL)-10, low IFN-γ, and no IL-4, IL-5, nor transforming growth factor (TGF)-β. The addition of anti–IL-10–neutralizing monoclonal antibodies during DC2 and CD8 T cell coculture, completely blocked the generation of IL-10–producing anergic CD8 T cells. IL-10–producing CD8 T cells strongly inhibit the allospecific proliferation of naive CD8 T cells to monocytes, and mature and immature DCs. This inhibition was mediated by IL-10, but not by TGF-β. IL-10–producing CD8 T cells could inhibit the bystander proliferation of naive CD8 T cells, provided that they were restimulated nearby to produce IL-10. IL-10–producing CD8 T cells could not inhibit the proliferation of DC1-preactivated effector T cells. This study demonstrates that IL-10–producing CD8 T cells are regulatory T cells, which provides a cellular basis for the phenomenon of CD8 T cell–mediated immunosuppression and suggests a role for plasmacytoid DC2 in immunological tolerance.


2018 ◽  
Vol 115 (18) ◽  
pp. 4749-4754 ◽  
Author(s):  
Eunseon Ahn ◽  
Koichi Araki ◽  
Masao Hashimoto ◽  
Weiyan Li ◽  
James L. Riley ◽  
...  

PD-1 (programmed cell death-1) is the central inhibitory receptor regulating CD8 T cell exhaustion during chronic viral infection and cancer. Interestingly, PD-1 is also expressed transiently by activated CD8 T cells during acute viral infection, but the role of PD-1 in modulating T cell effector differentiation and function is not well defined. To address this question, we examined the expression kinetics and role of PD-1 during acute lymphocytic choriomeningitis virus (LCMV) infection of mice. PD-1 was rapidly up-regulated in vivo upon activation of naive virus-specific CD8 T cells within 24 h after LCMV infection and in less than 4 h after peptide injection, well before any cell division had occurred. This rapid PD-1 expression by CD8 T cells was driven predominantly by antigen receptor signaling since infection with a LCMV strain with a mutation in the CD8 T cell epitope did not result in the increase of PD-1 on antigen-specific CD8 T cells. Blockade of the PD-1 pathway using anti–PD-L1 or anti–PD-1 antibodies during the early phase of acute LCMV infection increased mTOR signaling and granzyme B expression in virus-specific CD8 T cells and resulted in faster clearance of the infection. These results show that PD-1 plays an inhibitory role during the naive-to-effector CD8 T cell transition and that the PD-1 pathway can also be modulated at this stage of T cell differentiation. These findings have implications for developing therapeutic vaccination strategies in combination with PD-1 blockade.


Sign in / Sign up

Export Citation Format

Share Document