Attenuation of Immune Suppression to Complement Glioma Immunotherapy

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>


Genes ◽  
2021 ◽  
Vol 12 (6) ◽  
pp. 813
Author(s):  
Norwin Kubick ◽  
Pavel Klimovich ◽  
Patrick Henckell Flournoy ◽  
Irmina Bieńkowska ◽  
Marzena Łazarczyk ◽  
...  

Understanding the evolution of interleukins and interleukin receptors is essential to control the function of CD4+ T cells in various pathologies. Numerous aspects of CD4+ T cells’ presence are controlled by interleukins including differentiation, proliferation, and plasticity. CD4+ T cells have emerged during the divergence of jawed vertebrates. However, little is known about the evolution of interleukins and their origin. We traced the evolution of interleukins and their receptors from Placozoa to primates. We performed phylogenetic analysis, ancestral reconstruction, HH search, and positive selection analysis. Our results indicated that various interleukins' emergence predated CD4+ T cells divergence. IL14 was the most ancient interleukin with homologs in fungi. Invertebrates also expressed various interleukins such as IL41 and IL16. Several interleukin receptors also appeared before CD4+ T cells divergence. Interestingly IL17RA and IL17RD, which are known to play a fundamental role in Th17 CD4+ T cells first appeared in mollusks. Furthermore, our investigations showed that there is not any single gene family that could be the parent group of interleukins. We postulate that several groups have diverged from older existing cytokines such as IL4 from TGFβ, IL10 from IFN, and IL28 from BCAM. Interleukin receptors were less divergent than interleukins. We found that IL1R, IL7R might have diverged from a common invertebrate protein that contained TIR domains, conversely, IL2R, IL4R and IL6R might have emerged from a common invertebrate ancestor that possessed a fibronectin domain. IL8R seems to be a GPCR that belongs to the rhodopsin-like family and it has diverged from the Somatostatin group. Interestingly, several interleukins that are known to perform a critical function for CD4+ T cells such as IL6, IL17, and IL1B have gained new functions and evolved under positive selection. Overall evolution of interleukin receptors was not under significant positive selection. Interestingly, eight interleukin families appeared in lampreys, however, only two of them (IL17B, IL17E) evolved under positive selection. This observation indicates that although lampreys have a unique adaptive immune system that lacks CD4+ T cells, they could be utilizing interleukins in homologous mode to that of the vertebrates' immune system. Overall our study highlights the evolutionary heterogeneity within the interleukins and their receptor superfamilies and thus does not support the theory that interleukins evolved solely in jawed vertebrates to support T cell function. Conversely, some of the members are likely to play conserved functions in the innate immune system.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A929-A930
Author(s):  
Victoria Smith ◽  
Sterling Eckard ◽  
Bianca Rojo ◽  
Patrick Chun

BackgroundMDSC produce numerous immune-suppressive factors and are associated with poor outcomes across different cancers. They are frequently elevated in patients experiencing inadequate benefit from checkpoint blockade and there is a crucial need for therapies for this patient population. MDSC are recruited from bone marrow in response to both tumor signaling and T cell activation, and their accumulation in tumors and lymphatics can limit the potential benefits of immunostimulatory therapies. AMV564 is a bivalent T cell engager that selectively depletes MDSC. In a phase 1 study, pharmacodynamic analyses revealed significant depletion of MDSC, T cell activation, expansion of the T cell repertoire and an IFN-gamma-dominant cytokine profile with comparatively limited IL6 induction.1 Monotherapy activity including a confirmed RECIST complete response was observed. The clinical and pharmacodynamic profiles of AMV564 are being further evaluated in specific patient cohorts, including patients progressing on checkpoint blockade.MethodsIn a phase 1b expansion study (NCT04128423), patient cohorts with cancers more likely to include actionable tumor antigens were selected for treatment with AMV564, with most patients representing checkpoint treatment failures. An additional cohort of patients included heterogeneous tumor types stratified by tumor mutation burden (TMB) score from circulating tumor DNA. Pharmacodynamic analyses including direct immunophenotyping (flow cytometry) of T and myeloid cell compartments in peripheral blood were performed on patients treated with AMV564 (15 µg daily for 10 of 21 days by subcutaneous injection).ResultsChanges in myeloid and T cell profiles consistent with the pharmacodynamic signature of AMV564 were observed in patients receiving AMV564 despite one or more prior lines of checkpoint blockade therapy. Notably, both high baseline MDSC and elevated induction of MDSC after T cell activation were apparent (figure 1). Control of MDSC by AMV564 was associated with increases in both effector CD8 and CD4 T cells (figure 2). Extremely elevated levels of regulatory T cells were often observed: after treatment with AMV564, a Th-1-like repolarization of these cells was apparent, often associated with reduction in CD25 (figure 3).Abstract 887 Figure 1Significantly higher induction of M-MDSC is apparent in patients previously receiving checkpoint blockade (CPB) after T cell activation by AMV564.Abstract 887 Figure 2Treatment with AMV564 promotes increases in effector CD8 and CD4 T cells in patients previously treated with CPB (examples shown are Merkel cell carcinoma (MCC) and head and neck squamous cell carcinoma (HNSCC)).Abstract 887 Figure 3Th-1 like repolarization of Treg is apparent in patients previously treated with CPB (MCC, HNSCC examples) after treatment with AMV564 (a). Example CD25 low and T-Bet high cells in HNSCC patient (arrow, b).ConclusionsTreatment with AMV564 yielded substantial reductions in MDSC and favorable polarization of CD8 and CD4 T cells, including Th1-like polarization of Treg. This signature was apparent in patients previously treated with checkpoint inhibitors, despite strong induction of MDSC in response to T cell activation, and high baseline levels (>20%) of Treg.Trial RegistrationNCT04128423ReferencesSmith V, Eckard S, Rettig MP, et al. AMV564, a bivalent, bispecific T-cell engager, depletes myeloid derived suppressor cells and activates T cells in cancer patients. Cancer Res 2020;80(16 Supplement):5699.Ethics ApprovalThis study was approved by the Institutional Review Board (IRB) or Independent Ethics Committee (IEC) at each participating institution (including Ohio State University, MD Anderson Cancer Center, Duke University, University of California Los Angeles, Advent Health, Christ Hospital). All participants gave informed consent for samples used to generate pharmacodynamic data. No sensitive of identifiable information is included.


2021 ◽  
Vol 118 (37) ◽  
pp. e2103444118
Author(s):  
Felipe Valença-Pereira ◽  
Qian Fang ◽  
Isabelle J. Marié ◽  
Emily L. Giddings ◽  
Karen A. Fortner ◽  
...  

Interleukin 6 (IL-6) is known to regulate the CD4 T cell function by inducing gene expression of a number of cytokines through activation of Stat3 transcription factor. Here, we reveal that IL-6 strengthens the mechanics of CD4 T cells. The presence of IL-6 during activation of mouse and human CD4 T cells enhances their motility (random walk and exploratory spread), resulting in an increase in travel distance and higher velocity. This is an intrinsic effect of IL-6 on CD4 T-cell fitness that involves an increase in mitochondrial Ca2+. Although Stat3 transcriptional activity is dispensable for this process, IL-6 uses mitochondrial Stat3 to enhance mitochondrial Ca2+-mediated motility of CD4 T cells. Thus, through a noncanonical pathway, IL-6 can improve competitive fitness of CD4 T cells by facilitating cell motility. These results could lead to alternative therapeutic strategies for inflammatory diseases in which IL-6 plays a pathogenic role.


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.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1413-1413
Author(s):  
Akiko Fukunaga ◽  
Takayuki Ishikawa ◽  
Takero Shindo ◽  
Sumiko Takao ◽  
Toshiyuki Hori ◽  
...  

Abstract One of the major problems following allogeneic stem cell transplantation (allo-SCT) is the inability to reconstitute an adequate immune system for an extended period. T-cell reconstitution is also delayed for years, especially in CD4+ T cells. In addition to impaired thymic function, shortened Naive T cell survival due to altered T cell homeostasis is reported to be responsible for delayed immune reconstitution. To further investigate the mechanisms of delayed immune recovery after allo-SCT, we focused on the frequencies of effector CD4+ T cells, because according to the previous reports, progressive linear differentiation model of CD4+ T cell predicts the accumulation of terminally differentiated effector cells when transition from naïve to memory T cells and memory to effector cells are accelerated. By flowcytometric analyses we confirmed that CD27−CD4+ T cells from allo-SCT recipients uniformly express CD95, with negative expression of CCR7 and CD62L. They also produce g-interferon (IFNg) in response to the immobilized anti-CD3 and soluble anti-CD28 stimulation, which is consistent with previous reports insisting that CD27−CD4+ T cells are functionally differentiated effector T cells. Measuring the ratio of CD27−CD4+ T cells among CD4+ T cells revealed that, although healthy donors and patients received allo-SCT within a year had comparable CD27+CD4+T-cell rate (90% vs. 83%, P=0.4436), significantly decreased rate was observed in patients transplanted more than 1 year before (55% vs. 83%, P=0.0005). The ratio of CD27+CD4+ T cells kept low during the first 5 years after allo-SCT, and then it slowly begun to increase. In addition, in patients who received stem cell grafts more than 1 year before, the ratio of CD27+CD4+ T cells were significantly higher in patients transplanted from HLA-matched siblings than in those received unrelated grafts (69% vs. 42%, P=0.0002). Other factors, such as stem cell source (BM or PBSC), patient age, and the presence of chronic GVHD did not influence the ratio of CD27+CD4+ T cells. To further investigate the characteristics of CD27−CD4+ T cells in post-transplant periods, peripheral CD4+ T cells from patients who had received allo-SCT more than 1 year before as well as healthy volunteers were sorted into CD27− and CD27+ fractions, stained with CFSE, and stimulated with immobilized anti-CD3 and soluble anti-CD28 antibodies. CD27−CD4+ T cells proliferated more vigorously at 3 days after stimulation, though after another 2-day culture, there was no difference in cell divisions between both cell groups. In addition, CD27+ cells from transplanted patients lost their expression more frequently than those from volunteers, while none of the CD27− cells stored its expression. The fact of one-way transition from CD27+ to CD27− also supported that CD27−CD4+ T cells are terminally differentiated T cells. The finding that the frequencies of CD27−CD4+ T cells begin to elevate at 1 year after allo-SCT indicates that T cells infused with allograft do not easily lose the surface expression of CD27, while T cells derived from donor’s stem cells do. Considering the fact that ratio of CD27−CD4+ T cells is much higher in recipients of unrelated grafts, and it gradually begin to decrease at 5 years after allo-SCT, the increased ratio of CD27−CD4+ T cells may reflect altered T cell homeostasis. The serial monitoring of the ratio of CD27−CD4+ T cells after allo-SCT may be useful in evaluating immune reconstitution status.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 591-591 ◽  
Author(s):  
Patricia Taylor ◽  
Angela Panoskaltsis-Mortari ◽  
Gordon Freeman ◽  
Arlene Sharpe ◽  
Randolph Noelle ◽  
...  

Abstract ICOS, a CD28/CTLA-4 family member, is expressed on activated T cells. ICOS Ligand, a B7 family member, is constitutively expressed on B cells, monocytes and some T cells. Through the use of blocking anti-ICOS mAb and ICOS deficient (−/−) mice, we found that ICOS:ICOSL interactions play an important role in GVHD and BM graft rejection. Anti-ICOS mAb (given d-1 to d28 post BMT) significantly delayed or reduced mortality at 2 different T cell doses in a full MHC-disparate GVHD model. ICOS−/− T cells led to delayed or reduced mortality at 3 different cell doses compared to wild-type T cells. ICOS−/− CD4+ or CD8+ T cells infused into class II- or class I-disparate recipients, respectively, revealed that ICOS:ICOSL interactions regulate both CD4+ and CD8+ T cell alloresponses. Anti-ICOS inhibited GVHD in a CD28-independent fashion. Anti-ICOS inhibited GVHD mediated by either stat 4−/− or stat 6−/− T cells indicating that the ICOS pathway regulates both Th2 and Th1-mediated GVHD. In contrast to blockade of the B7:CD28/CTLA-4, CD40L:CD40 or the OX40:OX40L pathway, anti-ICOS mAb inhibited GVHD even when delayed until d5 post BMT, a time when substantial T cell expansion has occurred. A TCR transgenic model of GVHD was used to further study effects of ICOS:ICOSL blockade. All CB6 F1 recipients of anti-host alloreactive 2C CD8+ and TEa CD4+ T cells succumbed to GVHD mortality by d18 after transfer of cells. In contrast, 88% of anti-ICOS-treated mice survived long-term. Evaluation of spleens early after transplant revealed that anti-ICOS mAb reduced the number of TEa CD4+ cells by 44% and 2C CD8+ cells by 83%. Green fluorescent protein (GFP) 2C CD8+ and GFP TEa CD4+ T cells were infused into irradiated CB6 F1 mice and irrelevant or anti-ICOS mAb was administered. Mice were imaged on d4, 7 and 12 after T cell transfer. By d7, pronounced infiltration of GFP+ cells was noted in the peripheral and mesenteric LN, spleen, Peyer’s patches (PP), skin, gingiva, liver, kidney, lung, ileum, and colon of GVHD control mice. In contrast, there were fewer GFP+ cells in the spleen, ileum, colon, kidney, lung, skin and gingiva of anti-ICOS-treated mice, although there was no decrease in GFP+ cells in LNs or PP. To study the role of host ICOS expression in BM graft rejection, wild-type or ICOS−/− mice were sublethally irradiated and given allogeneic BM and evaluated for donor chimerism at 6 weeks post BMT. Five of 10 wild type mice engrafted (ave − 26% donor) in contrast to all 10 of ICOS−/− mice (ave − 71% donor). Collectively, these data indicate that ICOS:ICOSL interactions play an important role in GVHD, whether mediated by CD4+ Th1 or Th2 T cells or CD8+ T cells. Importantly, blockade of ICOS:ICOSL after initiation of alloresponses inhibited GVHD, in contrast to blockade of other costimulatory pathways, suggesting that the ICOS pathway may be a novel therapeutic target in primed transplantation situations. Anti-ICOS interfered with expansion of donor T cells in the spleen early after transplant and reduced the number of effector cells in several GVHD target tissues. These data suggest this pathway may be indicated for therapeutic targeting for the inhibition of GVHD and BM graft rejection.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 588-588
Author(s):  
Karrune Woan ◽  
Fengdong Cheng ◽  
Hongwei Wang ◽  
Jennifer Rock-Klotz ◽  
Zi Wang ◽  
...  

Abstract Abstract 588 We recently defined a novel role of histone deacetylase 11 (HDAC11), the newest member of the HDAC family, as a negative regulator of IL-10 gene transcription in antigen-presenting cells (APCs).1 To better understand the role of HDAC11 gene expression in immune cells in vivo, we have utilized a BAC (Bacterial artificial chromosome) transgenic mouse in which the EGFP reporter gene was inserted downstream of the HDAC11 promoter region but immediately upstream of the HDAC11 coding sequence (TgHDAC11-EGFP mice).2 In the steady-state, macrophages and B-cells isolated from spleen of TgHDAC11-EGFP mice express low levels of HDAC11 as evidenced by a slight shift in EGFP fluorescence from background. In sharp contrast, we identified a discrete population (11.9%) of T-cells over-expressing HDAC11 as demonstrated both by flow cytometry for EGFP and by qRT-PCR for HDAC11, a majority of which were CD4+ T-cells. Sorting of this EGFP+, CD4+ T-cell population confirmed that the increased EGFP expression correlated with an increased HDAC11mRNA expression. Reminiscent of our prior data in APCs, the increased expression of HDAC11 in T-cells was also inversely correlated with IL-10mRNA expression. Further analyses revealed that in the absence of any stimulation or T-cell polarizing conditions, this EGFP positive population expressed significantly elevated levels of ROR-γt and IL-17 mRNA, markers specific for the TH17 subpopulation. Polarization of wild type CD4+ T-cells into functional TH17 cells was associated with reduction of HDAC11 expression, suggesting a potential role for HDAC11 in regulating T-cell function and/or activation, in particular within the TH17 subset. Further support for this regulatory role of HDAC11 has been provided by our additional findings that T-cells devoid of HDAC11 are indeed hyper-reactive in vitro and in in vivo models. 1. Villagra A, et al. Nat Immunol. 2009 Jan;10(1):92-100. 2. Gong S, et al. Nature. 2003 Oct 30;425(6961):917-25. Disclosures: No relevant conflicts of interest to declare.


2003 ◽  
Vol 197 (7) ◽  
pp. 861-874 ◽  
Author(s):  
Ye Zheng ◽  
Monika Vig ◽  
Jesse Lyons ◽  
Luk Van Parijs ◽  
Amer A. Beg

Signaling pathways involved in regulating T cell proliferation and survival are not well understood. Here we have investigated a possible role of the nuclear factor (NF)-κB pathway in regulating mature T cell function by using CD4+ T cells from p50−/− cRel−/− mice, which exhibit virtually no inducible κB site binding activity. Studies with these mice indicate an essential role of T cell receptor (TCR)-induced NF-κB in regulating interleukin (IL)-2 expression, cell cycle entry, and survival of T cells. Our results further indicate that NF-κB regulates TCR-induced expression of antiapoptotic Bcl-2 family members. Strikingly, retroviral transduction of CD4+ T cells with the NF-κB–inducing IκB kinase β showed that NF-κB activation is not only necessary but also sufficient for T cell survival. In contrast, our results indicate a lack of involvement of NF-κB in both IL-2 and Akt-induced survival pathways. In vivo, p50−/− cRel−/− mice showed impaired superantigen-induced T cell responses as well as decreased numbers of effector/memory and regulatory CD4+ T cells. These findings provide the first demonstration of a role for NF-κB proteins in regulating T cell function in vivo and establish a critically important function of NF-κB in TCR-induced regulation of survival.


Sign in / Sign up

Export Citation Format

Share Document