scholarly journals A262 DIFFERENTIAL PHENOTYPES AND LOCALIZATION OF CD8+ T CELL RESPONSES TO ACUTE AND CHRONIC ENTERIC VIRAL INFECTION

2020 ◽  
Vol 3 (Supplement_1) ◽  
pp. 139-140
Author(s):  
B K Hardman ◽  
L C Osborne

Abstract Background Human Norovirus infection is the most common viral cause of gastroenteritis globally and the second most reported viral infection in Canada after the common cold. Most infections are acute, symptomatic, and rapidly cleared but some cases persist asymptomatically or induce post-infectious irritable bowel syndrome. Despite the global burden of these infections, no vaccine to prevent disease exists nor is the mechanism for persistence understood. MNV-CW3 and MNV-CR6 are murine noroviruses which demonstrate distinct biological behaviors that correlate with differential quantity and quality of antiviral CD8+ T cell responses. MNV-CW3 causes acute systemic infections initiated in the small intestine and cleared by day 8 due to a robust antiviral CD8+ T cell response. In contrast, MNV-CR6 causes chronic infections localized to the colonic intestinal epithelium and induces fewer antiviral CD8+ T cells with reduced effector molecule expression. Aims This research interrogates the mechanisms underlying strain-specific differential antiviral CD8+ T cell responses. Methods At days 3, 4, 5 and 8 post-infection, the phenotype and quantity of adoptively transferred MNV specific CD8+ T cells in the spleen, mesenteric lymph node (MLN), and the small and large intestine are analyzed by flow cytometry. Concurrently, immunofluorescent microscopy is used to determine whether CD8+ T cells are broadly disseminated throughout the intestines or localize in acute clusters of antiviral response. Combining these complementary techniques provides novel insight into mechanisms governing intestinal antiviral T cell responses. Results Activated MNV-specific CD8 T cells first accumulate in the MLN following oral infection with both MNV-CW3 and CR6, suggesting this is the site of immune activation. Supporting this hypothesis, preliminary data indicates that preventing T cell egress from activation sites by treatment with the S1PR1 agonist FTY720 leads to an enrichment of activated CD8+ T cells in the MLN following CW3 infection. Notably, the earliest stages of CD8+ T cell activation to CR6 infection is delayed compared to that elicited by CW3. Furthermore, at the peak of CD8+ T cell expansion (day 8 post-infection), CR6-elicited CD8+ T cells preferentially develop into short-lived effector populations rather than memory precursor populations. Conclusions These data reveal previously unknown differences in early events in CD8+ T cell activation following infection with two highly related viral strains that correlate with long-lasting effects on T cell differentiation and function. We are currently investigating the hypothesis that MNV-CW3 and CR6 may induce activation of distinct populations of, or pathways in, APC populations that would drive these differences. These results may have broad impacts on our understanding of how non-latent, chronic viral infections persist within a host. Funding Agencies CIHR

2014 ◽  
Vol 211 (2) ◽  
pp. 297-311 ◽  
Author(s):  
Danya Liu ◽  
Scott M. Krummey ◽  
I. Raul Badell ◽  
Maylene Wagener ◽  
Lumelle A. Schneeweis ◽  
...  

Mounting evidence in models of both autoimmunity and chronic viral infection suggests that the outcome of T cell activation is critically impacted by the constellation of co-stimulatory and co-inhibitory receptors expressed on the cell surface. Here, we identified a critical role for the co-inhibitory SLAM family member 2B4 (CD244) in attenuating primary antigen-specific CD8+ T cell responses in the presence of immune modulation with selective CD28 blockade. Our results reveal a specific up-regulation of 2B4 on antigen-specific CD8+ T cells in animals in which CD28 signaling was blocked. However, 2B4 up-regulation was not observed in animals treated with CTLA-4 Ig (abatacept) or CD28 blockade in the presence of anti–CTLA-4 mAb. 2B4 up-regulation after CD28 blockade was functionally significant, as the inhibitory impact of CD28 blockade was diminished when antigen-specific CD8+ T cells were deficient in 2B4. In contrast, 2B4 deficiency had no effect on CD8+ T cell responses during unmodified rejection or in the presence of CTLA-4 Ig. We conclude that blockade of CD28 signals in the presence of preserved CTLA-4 signals results in the unique up-regulation of 2B4 on primary CD8+ effectors, and that this 2B4 expression plays a critical functional role in controlling antigen-specific CD8+ T cell responses.


2012 ◽  
Vol 32 (suppl_1) ◽  
Author(s):  
Anna von Rossum ◽  
Winnie Enns ◽  
Yu P Shi ◽  
Jonathan C Choy

Transplant vasculopathy (TV) is an arteriosclerotic disease characterized by intimal thickening of allograft arteries and is a leading cause of heart transplant rejection. T cell responses towards allograft arteries are responsible for the development of TV and understanding the regulatory pathways controlling T cell activation in allograft arteries provides opportunities for the therapeutic attenuation of TV as well as other arteriosclerotic diseases. Bim is a pro-apoptotic Bcl-2 protein known to down-regulate immune responses after viral infections by inducing cell death of effector T cells but its role in regulating allogeneic T cell responses is not known. We compared cell death and alloantigen-driven activation of T cells from Bim +/+ (wild-type), Bim +/- and Bim -/- mice as well as the development of TV in these mice. Bim was required for cell death of both CD4 and CD8 T cells in response to cytokine deprivation in vitro . Unexpectedly, Bim was also required for alloantigen-induced proliferation of both CD4 and CD8 T cells as well as for IL-2 production. When TV was examined in aortic interposition grafts implanted into complete major histocompatibility complex-mismatched mice, intimal thickening was significantly reduced in Bim +/- but not Bim -/- recipients as compared to Bim +/+ counterparts. There was signficantly less CD4 T cell accumulation in the intima of arteries from Bim +/- as compared to Bim +/+ recipients but this effect was not observed in Bim -/- recipients. The accumulation of CD8 T cells in allograft arteries was not affected by differences in Bim expression. Taken together, our data support a novel role for Bim in driving T cell activation in response to allogeneic stimuli and indicate that the effects of this Bcl-2 protein in the pathogenesis of TV likely depends on its dual role in supporting T cell activation and death.


2021 ◽  
Author(s):  
◽  
Taryn Louise Osmond

<p>Splenic CD8α⁺ dendritic cells (DCs) have been described as key antigen presenting cells for the induction of CD8⁺ T cell responses to circulating antigen. This is through a heightened capacity to acquire and present the antigens via the process of cross-presentation, expression of high levels of the co-stimulatory and adhesion molecules required to stimulate CD8⁺ T cells, and the capacity to release high levels of the cytokines required to drive differentiation of CD8⁺ T cells into cytotoxic T lymphocytes (CTLs). However, recent research has indicated that the splenic CD8α⁺ DC population is more heterogeneous than originally thought. A previous study from my own laboratory suggested that a population of CD8α⁺ DCs that express the c-type lectin langerin primarily possess the heightened functions previously attributed to the total CD8α⁺ population. Therefore, the aim of this thesis research was to explore this subset of DCs in more detail, with specific emphasis on gaining mechanistic insight into their ability to elicit CD8⁺ T cell responses to circulating proteins. In the first section of this thesis, the hypothesis that the splenic langerin⁺ CD8α⁺ DCs were the critical subset involved in the induction of strong systemic CD8⁺ T cell responses to circulating antigen was tested in detail. This was examined using a genetically modified mouse model in which langerin-expressing cells could be easily identified and/or specifically depleted. It was first shown that the induction of CD8⁺ T cell responses to the model antigen ovalbumin was dependent on entry into the spleen in the presence of appropriate stimulation, which in these studies was provided by agonists for the toll-like receptors (TLRs) and/or signals from innate-like lymphocytes called natural killer T (NKT) cells. The primary targets for these signals were shown to be splenic langerin⁺ CD8α⁺ DCs, as CD8⁺ T cell responses were significantly reduced in hosts depleted of these cells within the spleen. Furthermore, agonists for TLRs that were not expressed by langerin⁺ CD8α⁺ DCs failed to enhance T cell responses. The langerin⁺ CD8α⁺ DCs were shown to be located in the marginal zone of the spleen, where they could readily screen the blood for antigens, and their function was critical to the induction of CD8⁺ T cell responses within six hours of antigen delivery. Interestingly, other local langerin-negative antigen presenting cells (APCs) were shown to be capable of cross-presentation, but with significantly reduced capacity to prime CD8⁺ T cell responses. Therefore, in the second section of this thesis the hypothesis that the langerin-negative APCs were capable of contributing to CD8⁺ T cell responses with appropriately timed stimuli was investigated. One of the downstream effects of inducing NKT cell activation at the time of priming was shown to be the “pre-conditioning” of langerin-negative DCs, allowing them to respond strongly to subsequent TLR ligation. Using SiglecH-DTR mice, it was shown that plasmacytoid DCs (which are langerin-negative) were pre-conditioned by NKT cell activation, allowing them to respond more actively to the delayed TLR stimulation by producing significantly enhanced levels of IFN-α. This factor was also potentially responsible for “feeding back” to the CD8α⁺ DCs (including langerin-expressing CD8α⁺ DCs), to enhance their function, as indicated by increases in cytokine production. Significantly, the major langerin-negative DC populations, defined as CD8α⁻ DCs, were pre-conditioned to have an enhanced cytokine release response to subsequent stimulation through TLR7, a receptor not expressed by langerin-positive DCs. This enhanced ability to respond to TLR7 ligation permitted these langerin-negative APCs to contribute to increased CD8⁺ T cell accumulation, with enhanced functional activity. Importantly, the CD8⁺ T cell response induced remained significantly dependent on initial cross-priming by langerin⁺ CD8α⁺ DCs, and it was only through pre-conditioning that langerinnegative APCs could contribute to enhancing the T cell response. In the third section of this thesis, the hypothesis that the CD8⁺ T cell responses generated in the presence of langerin⁺ CD8α⁺ DCs were phenotypically and functionally distinct from those responses generated in their absence was tested. No obvious differences were seen in CD8⁺ T cell homing, memory phenotype, restimulatory capacity, and expression of key molecules involved in metabolic function, survival and cytolytic function. However, in vivo cytotoxic function several weeks after priming was comparable, suggesting that this function was not related to initial burst size, providing some evidence of difference in function between CD8⁺ T cells primed in the presence or absence of langerin⁺ CD8α⁺ DCs. In summary, the splenic langerin⁺ CD8α⁺ DCs are the major subset responsible for cross-priming CD8⁺ T cell responses to circulating antigen, and for interpreting multiple stimulatory signals for enhancing the response. However, effective CD8⁺ T cell responses can be generated in their absence, particularly when antigens are provided in the context of appropriately temporally phased stimuli.</p>


2005 ◽  
Vol 201 (7) ◽  
pp. 1053-1059 ◽  
Author(s):  
Jason K. Whitmire ◽  
Joyce T. Tan ◽  
J. Lindsay Whitton

Interferon-γ (IFNγ) is important in regulating the adaptive immune response, and most current evidence suggests that it exerts a negative (proapoptotic) effect on CD8+ T cell responses. We have developed a novel technique of dual adoptive transfer, which allowed us to precisely compare, in normal mice, the in vivo antiviral responses of two T cell populations that differ only in their expression of the IFNγ receptor. We use this technique to show that, contrary to expectations, IFNγ strongly stimulates the development of CD8+ T cell responses during an acute viral infection. The stimulatory effect is abrogated in T cells lacking the IFNγ receptor, indicating that the cytokine acts directly upon CD8+ T cells to increase their abundance during acute viral infection.


2021 ◽  
Author(s):  
Takushi Nomura ◽  
Hiroyuki Yamamoto ◽  
Masako Nishizawa ◽  
Trang Hau ◽  
Shigeyoshi Harada ◽  
...  

SARS-CoV-2 infection presents clinical manifestations ranging from asymptomatic to fatal respiratory failure. Despite the induction of functional SARS-CoV-2-specific CD8+ T-cell responses in convalescent individuals, the role of virus-specific CD8+ T-cell responses in the control of SARS-CoV-2 replication remains unknown. In the present study, we show that subacute SARS-CoV-2 replication can be controlled in the absence of CD8+ T cells in cynomolgus macaques. Eight macaques were intranasally inoculated with 105 or 106 TCID50 of SARS-CoV-2, and three of the eight macaques were treated with a monoclonal anti-CD8 antibody on days 5 and 7 post-infection. In these three macaques, CD8+ T cells were undetectable on day 7 and thereafter, while virus-specific CD8+ T-cell responses were induced in the remaining five untreated animals. Viral RNA was detected in nasopharyngeal swabs for 10-17 days post-infection in all macaques, and the kinetics of viral RNA levels in pharyngeal swabs and plasma neutralizing antibody titers were comparable between the anti-CD8 antibody treated and untreated animals. SARS-CoV-2 RNA was detected in the pharyngeal mucosa and/or retropharyngeal lymph node obtained at necropsy on day 21 in two of the untreated group but undetectable in all macaques treated with anti-CD8 antibody. CD8+ T-cell responses may contribute to viral control in SARS-CoV-2 infection, but our results indicate possible containment of subacute viral replication in the absence of CD8+ T cells, implying that CD8+ T-cell dysfunction may not solely lead to viral control failure.


2021 ◽  
Author(s):  
◽  
Taryn Louise Osmond

<p>Splenic CD8α⁺ dendritic cells (DCs) have been described as key antigen presenting cells for the induction of CD8⁺ T cell responses to circulating antigen. This is through a heightened capacity to acquire and present the antigens via the process of cross-presentation, expression of high levels of the co-stimulatory and adhesion molecules required to stimulate CD8⁺ T cells, and the capacity to release high levels of the cytokines required to drive differentiation of CD8⁺ T cells into cytotoxic T lymphocytes (CTLs). However, recent research has indicated that the splenic CD8α⁺ DC population is more heterogeneous than originally thought. A previous study from my own laboratory suggested that a population of CD8α⁺ DCs that express the c-type lectin langerin primarily possess the heightened functions previously attributed to the total CD8α⁺ population. Therefore, the aim of this thesis research was to explore this subset of DCs in more detail, with specific emphasis on gaining mechanistic insight into their ability to elicit CD8⁺ T cell responses to circulating proteins. In the first section of this thesis, the hypothesis that the splenic langerin⁺ CD8α⁺ DCs were the critical subset involved in the induction of strong systemic CD8⁺ T cell responses to circulating antigen was tested in detail. This was examined using a genetically modified mouse model in which langerin-expressing cells could be easily identified and/or specifically depleted. It was first shown that the induction of CD8⁺ T cell responses to the model antigen ovalbumin was dependent on entry into the spleen in the presence of appropriate stimulation, which in these studies was provided by agonists for the toll-like receptors (TLRs) and/or signals from innate-like lymphocytes called natural killer T (NKT) cells. The primary targets for these signals were shown to be splenic langerin⁺ CD8α⁺ DCs, as CD8⁺ T cell responses were significantly reduced in hosts depleted of these cells within the spleen. Furthermore, agonists for TLRs that were not expressed by langerin⁺ CD8α⁺ DCs failed to enhance T cell responses. The langerin⁺ CD8α⁺ DCs were shown to be located in the marginal zone of the spleen, where they could readily screen the blood for antigens, and their function was critical to the induction of CD8⁺ T cell responses within six hours of antigen delivery. Interestingly, other local langerin-negative antigen presenting cells (APCs) were shown to be capable of cross-presentation, but with significantly reduced capacity to prime CD8⁺ T cell responses. Therefore, in the second section of this thesis the hypothesis that the langerin-negative APCs were capable of contributing to CD8⁺ T cell responses with appropriately timed stimuli was investigated. One of the downstream effects of inducing NKT cell activation at the time of priming was shown to be the “pre-conditioning” of langerin-negative DCs, allowing them to respond strongly to subsequent TLR ligation. Using SiglecH-DTR mice, it was shown that plasmacytoid DCs (which are langerin-negative) were pre-conditioned by NKT cell activation, allowing them to respond more actively to the delayed TLR stimulation by producing significantly enhanced levels of IFN-α. This factor was also potentially responsible for “feeding back” to the CD8α⁺ DCs (including langerin-expressing CD8α⁺ DCs), to enhance their function, as indicated by increases in cytokine production. Significantly, the major langerin-negative DC populations, defined as CD8α⁻ DCs, were pre-conditioned to have an enhanced cytokine release response to subsequent stimulation through TLR7, a receptor not expressed by langerin-positive DCs. This enhanced ability to respond to TLR7 ligation permitted these langerin-negative APCs to contribute to increased CD8⁺ T cell accumulation, with enhanced functional activity. Importantly, the CD8⁺ T cell response induced remained significantly dependent on initial cross-priming by langerin⁺ CD8α⁺ DCs, and it was only through pre-conditioning that langerinnegative APCs could contribute to enhancing the T cell response. In the third section of this thesis, the hypothesis that the CD8⁺ T cell responses generated in the presence of langerin⁺ CD8α⁺ DCs were phenotypically and functionally distinct from those responses generated in their absence was tested. No obvious differences were seen in CD8⁺ T cell homing, memory phenotype, restimulatory capacity, and expression of key molecules involved in metabolic function, survival and cytolytic function. However, in vivo cytotoxic function several weeks after priming was comparable, suggesting that this function was not related to initial burst size, providing some evidence of difference in function between CD8⁺ T cells primed in the presence or absence of langerin⁺ CD8α⁺ DCs. In summary, the splenic langerin⁺ CD8α⁺ DCs are the major subset responsible for cross-priming CD8⁺ T cell responses to circulating antigen, and for interpreting multiple stimulatory signals for enhancing the response. However, effective CD8⁺ T cell responses can be generated in their absence, particularly when antigens are provided in the context of appropriately temporally phased stimuli.</p>


2002 ◽  
Vol 76 (8) ◽  
pp. 3943-3951 ◽  
Author(s):  
M. Suresh ◽  
Gibson Lanier ◽  
Mary Katherine Large ◽  
Jason K. Whitmire ◽  
John D. Altman ◽  
...  

ABSTRACT The importance of lymphotoxin α (LTα) in lymphoid organogenesis is well established. Although LTα has been implicated in the pathogenesis of T-cell-mediated immunopathologies, the requirement for LTα in T-cell activation and effector function in vivo is not well understood. To determine the role of LTα in T-cell activation in vivo, we compared the generation of antigen-specific T-cell responses between wild type (+/+) and LTα-deficient (LTα−/−) mice during an acute infection with lymphocytic choriomeningitis virus (LCMV). Our studies showed that LCMV-infected LTα−/− mice had a profound impairment in the activation and expansion of virus-specific CD8 T cells in the spleen, as determined by cytotoxicity assays, intracellular staining for gamma interferon, and staining with major histocompatibility complex class I tetramers. Further, the nonlymphoid organs of LTα−/− mice also contained substantially lower number of LCMV-specific CD8 T cells than those of +/+ mice. Greatly reduced virus-specific CD8 T-cell responses in LTα−/− mice led to a defect in LCMV clearance from the tissues. In comparison to that in +/+ mice, the activation of LCMV-specific CD4 T cells was also significantly attenuated in LTα−/− mice. Adoptive transfer experiments were conducted to determine if abnormal lymphoid architecture in LTα−/− mice caused the impairment in the activation of LCMV-specific T-cell responses. Upon adoptive transfer into +/+ mice, the activation and expansion of LCMV-specific LTα−/− T cells were restored to levels comparable to those of +/+ T cells. In a reciprocal cell transfer experiment, activation of +/+ T cells was significantly reduced upon transfer into LTα−/− mice. These results showed that impairment in the activation of LCMV-specific T cells in LTα−/− mice may be due to abnormal lymphoid architecture and not to an intrinsic defect in LTα−/− T cells.


2005 ◽  
Vol 79 (15) ◽  
pp. 9419-9429 ◽  
Author(s):  
Nicole E. Miller ◽  
Jennifer R. Bonczyk ◽  
Yumi Nakayama ◽  
M. Suresh

ABSTRACT Although it is well documented that CD8 T cells play a critical role in controlling chronic viral infections, the mechanisms underlying the regulation of CD8 T-cell responses are not well understood. Using the mouse model of an acute and chronic lymphocytic choriomeningitis virus (LCMV) infection, we have examined the relative importance of peripheral T cells and thymic emigrants in the elicitation and maintenance of CD8 T-cell responses. Virus-specific CD8 T-cell responses were compared between mice that were either sham thymectomized or thymectomized (Thx) at ∼6 weeks of age. In an acute LCMV infection, thymic deficiency did not affect either the primary expansion of CD8 T cells or the proliferative renewal and maintenance of virus-specific lymphoid and nonlymphoid memory CD8 T cells. Following a chronic LCMV infection, in Thx mice, although the initial expansion of CD8 T cells was normal, the contraction phase of the CD8 T-cell response was exaggerated, which led to a transient but striking CD8 T-cell deficit on day 30 postinfection. However, the virus-specific CD8 T-cell response in Thx mice rebounded quickly and was maintained at normal levels thereafter, which indicated that the peripheral T-cell repertoire is quite robust and capable of sustaining an effective CD8 T-cell response in the absence of thymic output during a chronic LCMV infection. Taken together, these findings should further our understanding of the regulation of CD8 T-cell homeostasis in acute and chronic viral infections and might have implications in the development of immunotherapy.


2006 ◽  
Vol 81 (2) ◽  
pp. 934-944 ◽  
Author(s):  
Markus Cornberg ◽  
Brian S. Sheridan ◽  
Frances M. Saccoccio ◽  
Michael A. Brehm ◽  
Liisa K. Selin

ABSTRACT Live vaccinia virus (VV) vaccination has been highly successful in eradicating smallpox. However, the mechanisms of immunity involved in mediating this protective effect are still poorly understood, and the roles of CD8 T-cell responses in primary and secondary VV infections are not clearly identified. By applying the concept of molecular mimicry to identify potential CD8 T-cell epitopes that stimulate cross-reactive T cells specific to lymphocytic choriomeningitis virus (LCMV) and VV, we identified after screening only 115 peptides two VV-specific immunogenic epitopes that mediated protective immunity against VV. An immunodominant epitope, VV-e7r130, did not generate cross-reactive T-cell responses to LCMV, and a subdominant epitope, VV-a11r198, did generate cross-reactive responses to LCMV. Infection with VV induced strong epitope-specific responses which were stable into long-term memory and peaked at the time virus was cleared, consistent with CD8 T cells assisting in the control of VV. Two different approaches, direct adoptive transfer of VV-e7r-specific CD8 T cells and prior immunization with a VV-e7r-expressing ubiquitinated minigene, demonstrated that memory CD8 T cells alone could play a significant role in protective immunity against VV. These studies suggest that exploiting cross-reactive responses between viruses may be a useful tool to complement existing technology in predicting immunogenic epitopes to large viruses, such as VV, leading to a better understanding of the role CD8 T cells play during these viral infections.


2008 ◽  
Vol 105 (38) ◽  
pp. 14567-14572 ◽  
Author(s):  
E. Hammarlund ◽  
A. Dasgupta ◽  
C. Pinilla ◽  
P. Norori ◽  
K. Fruh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document