scholarly journals CD4 T cells are required for maintenance of CD8 TRM cells and virus control in the brain of MCMV-infected newborn mice

2019 ◽  
Vol 208 (3-4) ◽  
pp. 487-494 ◽  
Author(s):  
Ilija Brizić ◽  
Lea Hiršl ◽  
Marko Šustić ◽  
Mijo Golemac ◽  
William J. Britt ◽  
...  
2012 ◽  
Vol 189 (2) ◽  
pp. 968-979 ◽  
Author(s):  
Ana Villegas-Mendez ◽  
Rachel Greig ◽  
Tovah N. Shaw ◽  
J. Brian de Souza ◽  
Emily Gwyer Findlay ◽  
...  

1989 ◽  
Vol 170 (3) ◽  
pp. 1045-1050 ◽  
Author(s):  
J A Richt ◽  
L Stitz ◽  
H Wekerle ◽  
R Rott

A homogeneous T cell line NM1 with Borna disease (BD) virus reactivity could be established. The NM1 cells have been characterized as CD4+ T cells. Adoptive transfer revealed that this MHC class II-restricted immune cell is responsible for the immunopathological effect leading to BD, a progressive meningoencephalomyelitis.


2008 ◽  
Vol 82 (24) ◽  
pp. 12172-12180 ◽  
Author(s):  
Đurđica Cekinović ◽  
Mijo Golemac ◽  
Ester Pernjak Pugel ◽  
Jelena Tomac ◽  
Luka Čičin-Šain ◽  
...  

ABSTRACT Human cytomegalovirus (HCMV) is the most frequent cause of congenital viral infections in humans and frequently leads to long-term central nervous system (CNS) abnormalities that include learning disabilities, microcephaly, and hearing loss. The pathogenesis of the CNS infection has not been fully elucidated and may arise as a result of direct damage of CMV-infected neurons or indirectly secondary to inflammatory response to infection. We used a recently established model of mouse CMV (MCMV) infection in newborn mice to analyze the contribution of humoral immunity to virus clearance from the brain. In brains of MCMV-infected newborn mice treated with immune serum, the titer of infectious virus was reduced below detection limit, whereas in the brains of mice receiving control (nonimmune) serum significant amounts of virus were recovered. Moreover, histopathological and immunohistological analyses revealed significantly less CNS inflammation in mice treated with immune serum. Treatment with MCMV-specific monoclonal antibodies also resulted in the reduction of virus titer in the brain. Recipients of control serum or irrelevant antibodies had more viral foci, marked mononuclear cell infiltrates, and prominent glial nodules in their brains than mice treated with immune serum or MCMV-specific antibodies. In conclusion, our data indicate that virus-specific antibodies have a protective role in the development of CNS pathology in MCMV-infected newborn mice, suggesting that antiviral antibodies may be an important component of protective immunological responses during CMV infection of the developing CNS.


2018 ◽  
Author(s):  
Carleigh A. O’Brien ◽  
Samantha J. Batista ◽  
Katherine M. Still ◽  
Tajie H. Harris

AbstractControl of chronic CNS infection with the parasiteToxoplasma gondiirequires an ongoing T cell response in the brain. Immunosuppressive cytokines are also important for preventing lethal immunopathology during chronic infection. To explore the loss of suppressive cytokine exclusively during the chronic phase of infection we blocked IL-10 receptor (IL-10R). Blockade was associated with widespread changes in the inflammatory response, including increased antigen presenting cell (APC) activation, expansion of CD4+ T cells, and increased neutrophil recruitment to the brain, consistent with previous reports. We then sought to identify regulatory mechanisms contributing to IL-10 production, focusing on ICOS (inducible T cell costimulator), a molecule that promotes IL-10 production in many systems. Unexpectedly, ICOS-ligand (ICOSL) blockade led to a local expansion of effector T cells in the inflamed brain without affecting IL-10 production or APC activation. Instead, we found that ICOSL blockade led to changes in T cells associated with their proliferation and survival. Specifically, we observed increased expression of IL-2 associated signaling molecules, including CD25, STAT5 phosphorylation, Ki67, and Bcl-2 in T cells in the brain. Interestingly, increases in CD25 and Bcl-2 were not observed following IL-10R blockade. Also unlike IL-10R blockade, ICOSL blockade led to an expansion of both CD8+ and CD4+ T cells in the brain, with no expansion of peripheral T cell populations or neutrophil recruitment to the brain Overall, these results suggest that IL-10 and ICOS differentially regulate T cell responses in the brain during chronicT. gondiiinfection.


1991 ◽  
Vol 173 (4) ◽  
pp. 793-800 ◽  
Author(s):  
G B Huffnagle ◽  
J L Yates ◽  
M F Lipscomb

The role of CD4+ and CD8+ T cells in mediating pulmonary clearance of a cryptococcal infection was investigated. Intratracheal inoculation of BALB/c and C.B-17 mice with a moderately virulent strain of Cryptococcus neoformans (52D) resulted in a pulmonary infection, which was cleared by a T cell-dependent mechanism. During this clearance, there was a significant influx of both CD4+ and CD8+ T cells into the lungs. Depletion of CD4+ T cells by injections of CD4-specific monoclonal antibody (mAb) prevented pulmonary clearance and also resulted in significant colonization of the brain and spleen of infected mice. CD4 depletion did not prevent the influx of CD8+ T cells into the lungs. Surprisingly, depletion of CD8+ T cells by mAb also ablated pulmonary clearance. CD8-depleted mice also had a small but significant increase in brain and spleen colony-forming unit compared to control mice by the end of the study. CD4+ T cell pulmonary influx was independent of the presence of CD8+ T cells. The lungs of T cell-depleted mice were examined histologically. CD4+ and CD8+ T cells each mediated a degree of inflammatory influx seen in the lungs of infected mice and raised the possibility that CD4+ and CD8+ T cells may synergize to generate the inflammatory response in the lungs. Numerous phagocytized but intact cryptococci were seen in the inflammatory foci of CD8-depleted mice but not in control or CD4-depleted mice. We propose that CD4+ T cells may recruit and activate effector phagocytes while CD8+ T cells predominantly function to lyse cryptococcus-laden unactivated phagocytes similar to the function of CD8+ T cells during listeria and mycobacteria infections.


2000 ◽  
Vol 165 (6) ◽  
pp. 3128-3135 ◽  
Author(s):  
Paul R. Walker ◽  
Thomas Calzascia ◽  
Valérie Schnuriger ◽  
Nathalie Scamuffa ◽  
Philippe Saas ◽  
...  

Author(s):  
Xueyuan Yu ◽  
Jiajia Lv ◽  
Jun Wu ◽  
Yong Chen ◽  
Fei Chen ◽  
...  

Hyperactivation of NLRP3 inflammasome contributes to the neuroinflammation in autoimmune disorders, but the underlying regulating mechanism remains to be elucidated. We here demonstrate that mice lacking thymic stromal lymphopoietin receptor gene (Tslpr-/-) exhibit significant decreases in experimental autoimmune encephalitis (EAE) score, reduced CD4+ T cells infiltration, and restored expression of myelin basic protein (MBP) in the brain after induction of EAE by injection of myelin oligodendrocyte glycoprotein35-55 (MOG35-55) . TSLPR signals through Janus Kinase 2 (JAK2) to activate NLRP3. Tslpr-/- mice of EAE show decreased phosphorylation of JAK2 and expression of NLRP3 in the brain. In wild type (WT) mice after induction of EAE, inhibition of JAK2 by ruxolitinib inflammatory and CD4+ cell infiltration, decreased expression of NLRP3, and restored BMP expression in the brain. Ruxolitinib also decreased levels of IL-1β and TSLP in brain of EAE mouse when compared to that without ruxolitinib treatment. Further results with NLRP3 inhibitor MCC950 in EAE mouse of WT verified the proinflammatory role of NLRP3 by showing decreased inflammatory cells and CD4+ T cells, restored MBP expression, and declined levels of IL-1β and TSLP in the brain. In patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis we found increased level of NLRP3 and IL-1β in CSF when compared to that in control subjects. These findings highlight TSLP as a prospective target for treating JAK2-NLRP3 axis-associated autoimmune inflammatory disorders.


2021 ◽  
Author(s):  
◽  
Cameron Field

<p>Glioblastoma Multiforme (GBM) is a malignant primary brain tumour with an extremely poor prognosis. Following surgical resection, radiotherapy, and concomitant and adjuvant chemotherapy, median survival is only 12-15 months. New therapeutic approaches are therefore desperately needed.  Accumulating evidence suggests that activated T cells are capable of selectively targeting and eliminating tumour cells, even in the brain, making vaccine-mediated immunotherapy a promising candidate for the treatment of brain cancers. However, cancer vaccination has generally been disappointing in the clinic, and is unlikely to bestow long-term survival unless suppressive mechanisms are overcome. Checkpoint blockade is a recent treatment modality that enhances naturally occurring T cell responses to cancer by relieving suppression mediated by immune checkpoints – molecular signals that prevent T cell function. While significant clinical responses are often seen, it is clear that most patients fail to respond to checkpoint blockade alone. Therefore, there is considerable interest in combining the different immunotherapeutic strategies, with vaccines providing an immunogenic stimulus to induce anti-tumour T cells, and checkpoint blockade to ensure T cell function is retained.  An orthotopic murine model of glioma was utilised to examine this form of combined treatment. Immune responses induced with a unique whole-cell vaccine that utilises the adjuvant properties of invariant natural killer T cells (iNKT cells) were able to resist tumour challenge, but failed to eradicate established tumours. When the vaccine was combined with blocking antibodies to the immune checkpoint molecule cytotoxic T lymphocyte antigen-4 (α-CTLA-4) regression of established intracranial tumours was observed, whereas α-CTLA-4 was ineffective as a monotherapy. In contrast, combining the vaccine with antibodies to programmed death-1 (α-PD-1) or lymphocyte activation gene-3 (LAG-3) failed to provide any survival advantage. This was despite α-PD-1 being effective against the same tumour implanted subcutaneously, suggesting efficacy in the orthotopic setting was limited by poor access of α-PD-1 to effector T cells within the brain.  The effective combination of vaccine and α-CTLA-4 was associated with enhanced proliferation and accumulation of T cells in the lymphoid tissues without any obvious changes in the adjuvant function of iNKT cells or altered numbers of regulatory T cells, suggesting recently primed T cells were the targets of checkpoint inhibition. While tumours regressing under this combined treatment were highly infiltrated with a variety of leukocytes, tumour eradication was strictly dependent on CD4⁺ T cells.  Further interrogation of the cell-types responsible for anti-tumour activity revealed that CD11b⁺ cells were required for therapy, although it remains to be established whether these cells were involved in T cell priming or served as anti-tumour effectors in their own right, possibly under the influence of activated CD4⁺ T cells. In addition, therapy was hampered, although not entirely eliminated, in hosts deficient in interferon-γ. Therapy was also reduced significantly, but not entirely, in hosts deficient in perforin. In vitro studies showed that restimulated splenocytes from animals that had received the combined therapy were able to kill glioma cells in a perforin and MHC-II dependent manner, suggesting that cytotoxic CD4⁺ T cells were important effector cells.  Overall, these results demonstrate that immunotherapeutic vaccination can be combined effectively with checkpoint blockade to induce effective immune responses against glioma. The immune response induced in combination with CTLA-4 blockade differs from many other cancer models, with a strict dependence on CD4⁺ T cells that can serve either as cytotoxic effector cells, or potentially as modulators of other accessory cells. Furthermore, the tumour location presents new challenges, with access of inhibitors to the brain, particularly important if immune checkpoints on intratumoural effector cells are to be targeted. In this context, strategies to improve access of checkpoint inhibitors like α-PD-1 and α-LAG3 to the brain warrant further investigation.</p>


2021 ◽  
Author(s):  
◽  
Cameron Field

<p>Glioblastoma Multiforme (GBM) is a malignant primary brain tumour with an extremely poor prognosis. Following surgical resection, radiotherapy, and concomitant and adjuvant chemotherapy, median survival is only 12-15 months. New therapeutic approaches are therefore desperately needed.  Accumulating evidence suggests that activated T cells are capable of selectively targeting and eliminating tumour cells, even in the brain, making vaccine-mediated immunotherapy a promising candidate for the treatment of brain cancers. However, cancer vaccination has generally been disappointing in the clinic, and is unlikely to bestow long-term survival unless suppressive mechanisms are overcome. Checkpoint blockade is a recent treatment modality that enhances naturally occurring T cell responses to cancer by relieving suppression mediated by immune checkpoints – molecular signals that prevent T cell function. While significant clinical responses are often seen, it is clear that most patients fail to respond to checkpoint blockade alone. Therefore, there is considerable interest in combining the different immunotherapeutic strategies, with vaccines providing an immunogenic stimulus to induce anti-tumour T cells, and checkpoint blockade to ensure T cell function is retained.  An orthotopic murine model of glioma was utilised to examine this form of combined treatment. Immune responses induced with a unique whole-cell vaccine that utilises the adjuvant properties of invariant natural killer T cells (iNKT cells) were able to resist tumour challenge, but failed to eradicate established tumours. When the vaccine was combined with blocking antibodies to the immune checkpoint molecule cytotoxic T lymphocyte antigen-4 (α-CTLA-4) regression of established intracranial tumours was observed, whereas α-CTLA-4 was ineffective as a monotherapy. In contrast, combining the vaccine with antibodies to programmed death-1 (α-PD-1) or lymphocyte activation gene-3 (LAG-3) failed to provide any survival advantage. This was despite α-PD-1 being effective against the same tumour implanted subcutaneously, suggesting efficacy in the orthotopic setting was limited by poor access of α-PD-1 to effector T cells within the brain.  The effective combination of vaccine and α-CTLA-4 was associated with enhanced proliferation and accumulation of T cells in the lymphoid tissues without any obvious changes in the adjuvant function of iNKT cells or altered numbers of regulatory T cells, suggesting recently primed T cells were the targets of checkpoint inhibition. While tumours regressing under this combined treatment were highly infiltrated with a variety of leukocytes, tumour eradication was strictly dependent on CD4⁺ T cells.  Further interrogation of the cell-types responsible for anti-tumour activity revealed that CD11b⁺ cells were required for therapy, although it remains to be established whether these cells were involved in T cell priming or served as anti-tumour effectors in their own right, possibly under the influence of activated CD4⁺ T cells. In addition, therapy was hampered, although not entirely eliminated, in hosts deficient in interferon-γ. Therapy was also reduced significantly, but not entirely, in hosts deficient in perforin. In vitro studies showed that restimulated splenocytes from animals that had received the combined therapy were able to kill glioma cells in a perforin and MHC-II dependent manner, suggesting that cytotoxic CD4⁺ T cells were important effector cells.  Overall, these results demonstrate that immunotherapeutic vaccination can be combined effectively with checkpoint blockade to induce effective immune responses against glioma. The immune response induced in combination with CTLA-4 blockade differs from many other cancer models, with a strict dependence on CD4⁺ T cells that can serve either as cytotoxic effector cells, or potentially as modulators of other accessory cells. Furthermore, the tumour location presents new challenges, with access of inhibitors to the brain, particularly important if immune checkpoints on intratumoural effector cells are to be targeted. In this context, strategies to improve access of checkpoint inhibitors like α-PD-1 and α-LAG3 to the brain warrant further investigation.</p>


2021 ◽  
pp. ji2000773
Author(s):  
Nazanin Ghazanfari ◽  
Julia L. Gregory ◽  
Sapna Devi ◽  
Daniel Fernandez-Ruiz ◽  
Lynette Beattie ◽  
...  
Keyword(s):  
T Cells ◽  

Sign in / Sign up

Export Citation Format

Share Document