Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Immunol ; 212(12): 1904-1912, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38668728

ABSTRACT

NK cells have been shown to exhibit inflammatory and immunoregulatory functions in a variety of healthy and diseased settings. In the context of chronic viral infection and cancer, distinct NK cell populations that inhibit adaptive immune responses have been observed. To understand how these cells arise and further characterize their immunosuppressive role, we examined in vitro conditions that could polarize human NK cells into an inhibitory subset. TGF-ß1 has been shown to induce regulatory T cells in vitro and in vivo; we therefore investigated if TGF-ß1 could also induce immunosuppressive NK-like cells. First, we found that TGF-ß1/IL-15, but not IL-15 alone, induced CD103+CD49a+ NK-like cells from peripheral blood NK cells, which expressed markers previously associated with inhibitory CD56+ innate lymphoid cells, including high expression of GITR and CD101. Moreover, supernatant from ascites collected from patients with ovarian carcinoma also induced CD103+CD49a+ NK-like cells in vitro in a TGF-ß-dependent manner. Interestingly, TGF-ß1/IL-15-induced CD103+CD56+ NK-like cells suppressed autologous CD4+ T cells in vitro by reducing absolute number, proliferation, and expression of activation marker CD25. Collectively, these findings provide new insight into how NK cells may acquire an inhibitory phenotype in TGF-ß1-rich environments.


Subject(s)
Interleukin-15 , Killer Cells, Natural , Transforming Growth Factor beta1 , Humans , Killer Cells, Natural/immunology , Interleukin-15/immunology , Interleukin-15/metabolism , Transforming Growth Factor beta1/metabolism , Female , Antigens, CD/metabolism , Antigens, CD/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Integrin alpha Chains/metabolism , Integrin alpha Chains/immunology , CD56 Antigen/metabolism , Cells, Cultured , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Activation/immunology
2.
Cell ; 144(4): 601-13, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21295337

ABSTRACT

Understanding the factors that impede immune responses to persistent viruses is essential in designing therapies for HIV infection. Mice infected with LCMV clone-13 have persistent high-level viremia and a dysfunctional immune response. Interleukin-7, a cytokine that is critical for immune development and homeostasis, was used here to promote immunity toward clone-13, enabling elucidation of the inhibitory pathways underlying impaired antiviral immune response. Mechanistically, IL-7 downregulated a critical repressor of cytokine signaling, Socs3, resulting in amplified cytokine production, increased T cell effector function and numbers, and viral clearance. IL-7 enhanced thymic output to expand the naive T cell pool, including T cells that were not LCMV specific. Additionally, IL-7 promoted production of cytoprotective IL-22 that abrogated liver pathology. The IL-7-mediated effects were dependent on endogenous IL-6. These attributes of IL-7 have profound implications for its use as a therapeutic in the treatment of chronic viral diseases.


Subject(s)
Interleukin-7/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Animals , Antigens, Differentiation/metabolism , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Interleukin-6/immunology , Interleukin-7/immunology , Mice , Programmed Cell Death 1 Receptor , Recombinant Proteins/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/immunology
3.
J Immunol ; 208(12): 2702-2712, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35667842

ABSTRACT

CD8+ T cell proliferation and differentiation into effector and memory states are high-energy processes associated with changes in cellular metabolism. CD28-mediated costimulation of T cells activates the PI3K/AKT/mammalian target of rapamycin signaling pathway and induces eukaryotic translation initiation factor 4E-dependent translation through the derepression by 4E-BP1 and 4E-BP2. In this study, we demonstrate that 4E-BP1/2 proteins are required for optimum proliferation of mouse CD8+ T cells and the development of an antiviral effector function. We show that translation of genes encoding mitochondrial biogenesis is impaired in T cells derived from 4E-BP1/2-deficient mice. Our findings demonstrate an unanticipated role for 4E-BPs in regulating a metabolic program that is required for cell growth and biosynthesis during the early stages of CD8+ T cell expansion.


Subject(s)
Eukaryotic Initiation Factors , Phosphoproteins , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Mammals/genetics , Mice , Organelle Biogenesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis
4.
Cancer Immunol Immunother ; 68(5): 773-785, 2019 May.
Article in English | MEDLINE | ID: mdl-30747243

ABSTRACT

Adoptive cell therapy using autologous tumor-infiltrating lymphocytes (TIL) has shown significant clinical benefit, but is limited by toxicities due to a requirement for post-infusion interleukin-2 (IL-2), for which high dose is standard. To assess a modified TIL protocol using lower dose IL-2, we performed a single institution phase II protocol in unresectable, metastatic melanoma. The primary endpoint was response rate. Secondary endpoints were safety and assessment of immune correlates following TIL infusion. Twelve metastatic melanoma patients were treated with non-myeloablative lymphodepleting chemotherapy, TIL, and low-dose subcutaneous IL-2 (125,000 IU/kg/day, maximum 9-10 doses over 2 weeks). All but one patient had previously progressed after treatment with immune checkpoint inhibitors. No unexpected adverse events were observed, and patients received an average of 6.8 doses of IL-2. By RECIST v1.1, two patients experienced a partial response, one patient had an unconfirmed partial response, and six had stable disease. Biomarker assessment confirmed an increase in IL-15 levels following lymphodepleting chemotherapy as expected and a lack of peripheral regulatory T-cell expansion following protocol treatment. Interrogation of the TIL infusion product and monitoring of the peripheral blood following infusion suggested engraftment of TIL. In one responding patient, a population of T cells expressing a T-cell receptor Vß chain that was dominant in the infusion product was present at a high percentage in peripheral blood more than 2 years after TIL infusion. This study shows that this protocol of low-dose IL-2 following adoptive cell transfer of TIL is feasible and clinically active. (ClinicalTrials.gov identifier NCT01883323.).


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-2/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Cell Proliferation , Cells, Cultured , Female , Humans , Interleukin-15/metabolism , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma/immunology , Middle Aged , Neoplasm Metastasis , Skin Neoplasms/immunology , Treatment Outcome
5.
J Immunol ; 199(12): 4056-4065, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29109121

ABSTRACT

The decision between T cell activation and tolerance is governed by the spatial and temporal integration of diverse molecular signals and events occurring downstream of TCR and costimulatory or coinhibitory receptor engagement. The PI3K-protein kinase B (PKB; also known as Akt) signaling pathway is a central axis in mediating proximal signaling events of TCR and CD28 engagement in T cells. Perturbation of the PI3K-PKB pathway, or the loss of negative regulators of T cell activation, such as the E3 ubiquitin ligase Cbl-b, have been reported to lead to increased susceptibility to autoimmunity. In this study, we further examined the molecular pathway linking PKB and Cbl-b in murine models. Our data show that the protein kinase GSK-3, one of the first targets identified for PKB, catalyzes two previously unreported phosphorylation events at Ser476 and Ser480 of Cbl-b. GSK-3 inactivation by PKB abrogates phosphorylation of Cbl-b at these two sites and results in reduced Cbl-b protein levels. We further show that constitutive activation of PKB in vivo results in a loss of tolerance that is mediated through the downregulation of Cbl-b. Altogether, these data indicate that the PI3K-PKB-GSK-3 pathway is a novel regulatory axis that is important for controlling the decision between T cell activation and tolerance via Cbl-b.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glycogen Synthase Kinase 3/physiology , Immune Tolerance/physiology , Lymphocyte Activation/physiology , Proto-Oncogene Proteins c-cbl/metabolism , T-Lymphocyte Subsets/enzymology , Amino Acid Sequence , Animals , Autoimmunity/physiology , Enzyme Activation , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Phosphoserine/metabolism , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/physiology , Sequence Alignment , Signal Transduction/physiology , Species Specificity , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology
6.
J Immunol ; 190(3): 1210-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275599

ABSTRACT

Recent studies have begun to define the role of micro-RNAs in regulating the immune response. Micro-RNA155 (mir-155) has been shown to play a role in germinal center formation, T cell inflammation, and regulatory T cell development. In this study, we evaluated the role of mir-155 in cytotoxic T cell function. We report in this study that mice lacking mir-155 have impaired CD8(+) T cell responses to infections with lymphocytic choriomeningitis virus and the intracellular bacteria Listeria monocytogenes. We show by a series of adoptive transfer studies that the impaired CD8(+) T cell response to L. monocytogenes is T cell intrinsic. In addition, we observed that CD8(+) T cells lacking mir-155 have impaired activation of the prosurvival Akt pathway after TCR cross-linking. These data suggest that mir-155 may be a good target for therapies aimed at modulating immune responses.


Subject(s)
Listeriosis/immunology , Lymphocytic Choriomeningitis/immunology , MicroRNAs/physiology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Bacterial/immunology , Apoptosis/immunology , Clonal Selection, Antigen-Mediated , Cytotoxicity, Immunologic , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Peptide Fragments/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/transplantation
7.
J Immunol ; 191(4): 1704-15, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851691

ABSTRACT

Differentiation of CD8 single-positive (SP) T cells is predicated by the ability of lymphocyte progenitors to integrate multiple signaling cues provided by the thymic microenvironment. In the thymus and the OP9-DL1 system for T cell development, Notch signals are required for progenitors to commit to the T cell lineage and necessary for their progression to the CD4(+)CD8(+) double-positive (DP) stage of T cell development. However, it remains unclear whether Notch is a prerequisite for the differentiation of DP cells to the CD8 SP stage of development. In this study, we demonstrate that Notch receptor-ligand interactions allow for efficient differentiation and selection of conventional CD8 T cells from bone marrow-derived hematopoietic stem cells. However, bone marrow-derived hematopoietic stem cells isolated from Itk(-/-)Rlk(-/-) mice gave rise to T cells with decreased IFN-γ production, but gained the ability to produce IL-17. We further reveal that positive and negative selection in vitro are constrained by peptide-MHC class I expressed on OP9 cells. Finally, using an MHC class I-restricted TCR-transgenic model, we show that the commitment of DP precursors to the CD8 T cell lineage is dependent on Notch signaling. Our findings further establish the requirement for Notch receptor-ligand interactions throughout T cell differentiation, including the final step of CD8 SP selection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated , Intercellular Signaling Peptides and Proteins/immunology , Lymphopoiesis/immunology , Receptors, Notch/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Actins/immunology , Animals , Antigens, Viral/immunology , CD4 Antigens/analysis , CD8 Antigens/analysis , Calcium-Binding Proteins , Cell Lineage , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Crosses, Genetic , H-2 Antigens/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Histocompatibility Antigen H-2D/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Specific Pathogen-Free Organisms , Stromal Cells/cytology , Stromal Cells/immunology
8.
Proc Natl Acad Sci U S A ; 109(4): 1210-5, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22167808

ABSTRACT

Infections with HIV, hepatitis B virus, and hepatitis C virus can turn into chronic infections, which currently affect more than 500 million patients worldwide. It is generally thought that virus-mediated T-cell exhaustion limits T-cell function, thus promoting chronic disease. Here we demonstrate that natural killer (NK) cells have a negative impact on the development of T-cell immunity by using the murine lymphocytic choriomeningitis virus. NK cell-deficient (Nfil3(-/-), E4BP4(-/-)) mice exhibited a higher virus-specific T-cell response. In addition, NK cell depletion caused enhanced T-cell immunity in WT mice, which led to rapid virus control and prevented chronic infection in lymphocytic choriomeningitis virus clone 13- and reduced viral load in DOCILE-infected animals. Further experiments showed that NKG2D triggered regulatory NK cell functions, which were mediated by perforin, and limited T-cell responses. Therefore, we identified an important role of regulatory NK cells in limiting T-cell immunity during virus infection.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Communicable Diseases/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus , Analysis of Variance , Animals , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histological Techniques , Interferon-alpha/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily K/immunology , Real-Time Polymerase Chain Reaction
9.
Immunol Rev ; 241(1): 164-79, 2011 May.
Article in English | MEDLINE | ID: mdl-21488897

ABSTRACT

Self-reactive T cells in the body are controlled by mechanisms of peripheral tolerance that limit their activation and induction of immune pathology. Our understanding of these mechanisms has been advanced by the use of tissue-specific promoters to express neo-self-antigens. Here, we present findings using the RIP-gp (rat insulin promoter-glycoprotein) transgenic mouse, which expresses the lymphocytic choriomeningitis virus glycoprotein (LCMV-gp) specifically in the pancreatic ß islet cells. T cells responsive to this antigen remain ignorant of the LCMV-gp expressed by the islets, and breaking tolerance is dependent upon the maturation status of antigen-presenting cells, the avidity of the T-cell receptor ligation, and the level of major histocompatibility complex expression in the pancreas. Furthermore, decreased activity of Casitas B-lineage lymphoma b, a negative regulator of T-cell receptor signaling, can allow recognition and destruction of the pancreatic islets. This review discusses the roles of these factors in the context of anti-tissue responses, both in the setting of autoimmunity and in anti-tumor immunity.


Subject(s)
Autoantigens/metabolism , Autoimmune Diseases/immunology , Glycoproteins/metabolism , Insulin-Secreting Cells/metabolism , Neoplasms/immunology , Viral Envelope Proteins/metabolism , Animals , Antigens, Neoplasm/immunology , Autoantigens/genetics , Autoantigens/immunology , Glycoproteins/genetics , Humans , Insulin/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic/genetics , Self Tolerance , Transgenes/genetics , Viral Envelope Proteins/genetics
10.
Methods Cell Biol ; 188: 153-169, 2024.
Article in English | MEDLINE | ID: mdl-38880522

ABSTRACT

Pancreatic cancer remains an unmet medical need. Late diagnosis and the lack of efficient treatment significantly impact the prognosis of patients suffering from pancreatic cancer. Improving patient outcomes requires a deeper comprehension of the tumor ecosystem. To achieve this, a thorough exploration of the tumor microenvironment using pre-clinical models that accurately replicate human disease is imperative, particularly in understanding the dynamics of immune cell subsets. Surprisingly, the impact of model variations on the composition of the tumor microenvironment has been largely neglected. In this study, we introduce an orthotopic model of pancreatic ductal adenocarcinoma and a spontaneous model of insulinoma. Our findings reveal striking differences in the innate lymphoid cell infiltrate, highlighting the importance of considering model-specific influences when investigating the tumor microenvironment.


Subject(s)
Carcinoma, Pancreatic Ductal , Disease Models, Animal , Immunity, Innate , Lymphocytes , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Tumor Microenvironment/immunology , Lymphocytes/immunology , Humans , Insulinoma/pathology , Insulinoma/immunology , Cell Line, Tumor , Mice, Inbred C57BL
11.
Cell Rep Med ; 5(3): 101465, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38460518

ABSTRACT

The manipulation of T cell metabolism to enhance anti-tumor activity is an area of active investigation. Here, we report that activating the amino acid starvation response in effector CD8+ T cells ex vivo using the general control non-depressible 2 (GCN2) agonist halofuginone (halo) enhances oxidative metabolism and effector function. Mechanistically, we identified autophagy coupled with the CD98-mTOR axis as key downstream mediators of the phenotype induced by halo treatment. The adoptive transfer of halo-treated CD8+ T cells into tumor-bearing mice led to robust tumor control and curative responses. Halo-treated T cells synergized in vivo with a 4-1BB agonistic antibody to control tumor growth in a mouse model resistant to immunotherapy. Importantly, treatment of human CD8+ T cells with halo resulted in similar metabolic and functional reprogramming. These findings demonstrate that activating the amino acid starvation response with the GCN2 agonist halo can enhance T cell metabolism and anti-tumor activity.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Animals , Mice , Immunotherapy, Adoptive/methods , Neoplasms/pathology , Immunotherapy , Amino Acids
12.
Cancer Immunol Res ; 10(4): 437-452, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35181779

ABSTRACT

Regulatory T cells (Treg) are an integral component of the adaptive immune system that negatively affect antitumor immunity. Here, we investigated the role of the E3 ubiquitin ligase casitas B-lineage lymphoma-b (Cbl-b) in establishing CD8+ T-cell resistance to Treg-mediated suppression to enhance antitumor immunity. Transcriptomic analyses suggested that Cbl-b regulates pathways associated with cytokine signaling and cellular proliferation. We showed that the hypersecretion of IFNγ by Cbl-b-deficient CD8+ T cells selectively attenuated CD8+ T-cell suppression by Tregs. Although IFNγ production by Cbl-b-deficient T cells contributed to phenotypic alterations in Tregs, the cytokine did not attenuate the suppressive function of Tregs. Instead, IFNγ had a profound effect on CD8+ T cells by directly upregulating interferon-stimulated genes and modulating T-cell activation. In murine models of adoptive T-cell therapy, Cbl-b-deficient T cells elicited superior antitumor immune response. Furthermore, Cbl-b-deficient CD8+ T cells were less susceptible to suppression by Tregs in the tumor through the effects of IFNγ. Collectively, this study demonstrates that the hypersecretion of IFNγ serves as a key mechanism by which Cbl-b-deficient CD8+ T cells are rendered resistant to Tregs. See related Spotlight by Wolf and Baier, p. 370.


Subject(s)
T-Lymphocytes, Regulatory , Adaptor Proteins, Signal Transducing , Animals , CD8-Positive T-Lymphocytes , Interferon-gamma/metabolism , Mice , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism
13.
J Exp Med ; 202(3): 405-13, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16043518

ABSTRACT

Fas-associated death domain (FADD) and caspase-8 are key signal transducers for death receptor-induced apoptosis, whereas cellular FLICE-inhibitory protein (cFLIP) antagonizes this process. Interestingly, FADD and caspase-8 also play a role in T cell development and T cell receptor (TCR)-mediated proliferative responses. To investigate the underlying mechanism, we generated cFLIP-deficient T cells by reconstituting Rag-/- blastocysts with cFLIP-deficient embryonic stem cells. These Rag chimeric mutant mice (rcFLIP-/-) had severely reduced numbers of T cells in the thymus, lymph nodes, and spleen, although mature T lymphocytes did develop. Similar to FADD- or caspase-8-deficient cells, rcFLIP-/- T cells were impaired in proliferation in response to TCR stimulation. Further investigation revealed that cFLIP is required for T cell survival, as well as T cell cycling in response to TCR stimulation. Interestingly, some signaling pathways from the TCR complex appeared competent, as CD3 plus CD28 cross-linking was capable of activating the ERK pathway in rcFLIP-/- T cells. We demonstrate an essential role for cFLIP in T cell function.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/physiology , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein , CD28 Antigens/metabolism , CD3 Complex/metabolism , Caspase 8 , Caspases/metabolism , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas-Associated Death Domain Protein , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism
14.
Eur J Immunol ; 40(12): 3372-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21110320

ABSTRACT

T-cell death is a fundamental process that is intricately regulated at multiple phases during T-cell differentiation, tolerance induction and the decline of the immune response. Caspase 3 is a crucial molecule regulating both mitochondrial and death receptor apoptotic pathways and therefore we were interested in examining the role of caspase 3 in T cells. Using P14 and H-Y CD8(+) TCR-transgenic models, our analysis has shown that caspase 3 is not required for thymic negative selection. In addition, caspase 3 does not play a prominent role in the contraction phase following acute viral infection, nor clonal deletion of CD8(+) T cells under tolerizing conditions. Surprisingly, our studies demonstrate that caspase 3 was not required for the induction of CD8(+) T-cell anergy in vivo, contrary to published reports using CD4(+) T cells. Therefore, these results demonstrate that caspase 3 is not essential in CD8(+) T cells for multiple forms of thymic or peripheral tolerance, nor the contraction phase after an acute anti-viral response.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Caspase 3/metabolism , Clonal Anergy , Thymus Gland/cytology , Animals , Apoptosis/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Caspase 3/immunology , Cell Differentiation/immunology , Clonal Anergy/immunology , H-Y Antigen/immunology , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Signal Transduction/immunology , Thymus Gland/growth & development , Thymus Gland/immunology
15.
Eur J Immunol ; 40(3): 677-81, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20082358

ABSTRACT

Regulatory T (Treg) cells are crucial for maintaining peripheral tolerance and controlling T-cell responses. The generation of Treg in the thymus requires TCR triggering and CD28 costimulation. Engagement of these receptors induces a number of signalling pathways, including the activation of NF-kappaB via PKCtheta and the Bcl-10/CARMA1/MALT complex. Previous studies have shown that PKCtheta, Bcl-10 and CARMA1 are important for Treg development. It is unclear, however, whether different members of the NF-kappaB family contribute to Treg development or homeostasis. In this study, we show that Treg numbers are reduced in the absence of c-Rel but not NF-kappaB1 (p50). Furthermore, using mixed bone marrow chimeras from WT and KO animals, we demonstrate that the requirement for PKCtheta, Bcl-10 and c-Rel is T-cell intrinsic, and cannot be rescued by the presence of WT cells. Therefore, c-Rel and NF-kappaB1 have differential roles in Treg development.


Subject(s)
Cell Differentiation/immunology , NF-kappa B/immunology , Proto-Oncogene Proteins c-rel/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/immunology
16.
Eur J Immunol ; 40(3): 867-77, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19950170

ABSTRACT

Elucidating the signaling events that promote T-cell tolerance versus activation provides important insights for manipulating immunity in vivo. Previous studies have suggested that the absence of PKCtheta results in the induction of anergy and that the balance between the induction of the transcription factors NFAT, AP1 and NF-kappaB plays a key role in determining whether T-cell anergy or activation is induced. Here, we examine whether Bcl-10 and specific family members of NF-kappaB act downstream of PKCtheta to alter CD8(+) T-cell activation and/or anergy. We showed that T cells from mice deficient in c-Rel but not NF-kappaB1 (p50) have increased susceptibility to the induction of anergy, similar to T cells from PKCtheta-deficient mice. Surprisingly T cells from Bcl-10-deficient mice showed a strikingly different phenotype to the PKCtheta-deficient T cells, with a severe block in TCR-mediated activation. Furthermore, we have also shown that survival signals downstream of NF-kappaB, are uncoupled from signals that mediate T-cell anergy. These results suggest that c-Rel plays a critical role downstream of PKCtheta in controlling CD8(+) T-cell anergy induction.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Isoenzymes/immunology , Lymphocyte Activation/immunology , Protein Kinase C/immunology , Proto-Oncogene Proteins c-rel/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein , Blotting, Western , Mice , Mice, Transgenic , NF-kappa B/immunology , Phenotype , Protein Kinase C-theta , Signal Transduction/immunology
17.
J Immunol ; 183(9): 5728-37, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812207

ABSTRACT

Inflammation following tissue damage promotes lymphocyte recruitment, tissue remodeling, and wound healing while maintaining self tolerance. Endogenous signals associated with tissue damage and cell death have been proposed to initiate and instruct immune responses following injury. In this study, we have examined the effects of elevated levels of a candidate endogenous danger signal, heat shock cognate protein 70 (hsc70), on stimulation of inflammation and autoimmunity following cell damage. We find that damage to pancreatic beta cells expressing additional cytosolic hsc70 leads to an increased incidence of diabetes in a transgenic mouse model. Steady-state levels of activated APC and T cell populations in the draining lymph node were enhanced, which further increased following streptozotocin-induced beta cell death. In addition, proinflammatory serum cytokines, and lymphocyte recruitment were increased in hsc70 transgenic mice. Islet Ag-specific T cells underwent a greater extent of proliferation in the lymph nodes of mice expressing hsc70 following beta cell damage, suggesting elevated Ag presentation following release of Ag in the presence of hsc70. These findings suggest that an elevated content of hsc70 in cells undergoing necrotic or apoptotic cell death can increase the extent of sterile inflammation and increase the susceptibility to autoimmunity.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , HSC70 Heat-Shock Proteins/biosynthesis , HSC70 Heat-Shock Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Animals , Autoimmune Diseases/epidemiology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cattle , Cell Death/genetics , Cell Death/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytosol/immunology , Cytosol/metabolism , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Incidence , Islets of Langerhans/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Rats
18.
Nat Med ; 9(12): 1469-76, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625545

ABSTRACT

Pathogens or pathogen-associated molecular patterns can signal to cells of the innate immune system and trigger effective adaptive immunity. However, relatively little is known about how the innate immune system detects tissue injury or necrosis. Evidence suggests that the release of heat-shock proteins (HSPs) may provide adjuvant-like signals, but the ability of HSPs to promote activation or tolerance in vivo has not been addressed. In this study we show that Hsp70 promotes dendritic cell (DC) function and, together with antigen, triggers autoimmune disease in vivo.


Subject(s)
Antigen-Presenting Cells/immunology , HSP70 Heat-Shock Proteins/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity , Base Sequence , CD40 Antigens/metabolism , DNA, Complementary/genetics , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Humans , Immune Tolerance , In Vitro Techniques , Interleukin-12/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/immunology , Signal Transduction
19.
Nat Med ; 10(11): 1234-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15467726

ABSTRACT

Autoimmune diseases are often mediated by self-reactive T cells, which must be activated to cause immunopathology. One mechanism, known as molecular mimicry, proposes that self-reactive T cells may be activated by pathogens expressing crossreactive ligands. Here we have developed a model to investigate how the affinity of the T-cell receptor (TCR) for the activating agent influences autoimmunity. Our model shows that an approximately fivefold difference in the TCR affinity for the activating ligand results in a 50% reduction in the incidence of autoimmunity. A reduction in TCR-ligand affinity to approximately 20 times lower than normal does not induce autoimmunity despite the unexpected induction of cytotoxic T lymphocytes (CTLs) and insulitis. Furthermore, in the absence of a key negative regulatory molecule, Cbl-b, 100% of mice develop autoimmunity upon infection with viruses encoding the lower-affinity ligand. Therefore, autoimmune disease is sensitive both to the affinity of the activating ligand and to normal mechanisms that negatively regulate the immune response.


Subject(s)
Autoimmunity/immunology , Models, Immunological , Molecular Mimicry/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , Cell Proliferation , Chromium Radioisotopes , Diabetes Mellitus, Experimental/immunology , Flow Cytometry , Humans , Immunohistochemistry , Ligands , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/immunology
20.
Proc Natl Acad Sci U S A ; 105(8): 2999-3004, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287017

ABSTRACT

There are many inhibitory mechanisms that function at the cellular and molecular levels to maintain tolerance. Despite this, self-reactive clones escape regulatory mechanisms and cause autoimmunity in certain circumstances. We hypothesized that the same mechanisms that permit T cells to expand during homeostatic proliferation may inadvertently promote autoimmunity under certain conditions. One major homeostatic cytokine is IL-7, and studies have linked it or its receptor to the development of multiple sclerosis and other autoimmune diseases. We show in a model of beta-islet cell self-reactivity that the transfer of activated autoreactive CD4 T cells can prime and expand endogenous autoreactive CD8 T cells in a CD28- and CD40-dependent manner through the licensing of dendritic cells. Despite this, mice do not develop diabetes. However, the provision of exogenous IL-7 or the physiological production of IL-7 associated with lymphopenia was able to profoundly promote the expansion of self-reactive clones even in the presence of regulatory T cells. Autoimmune diabetes rapidly ensued with CD4 help and the subsequent activation of CD8 T cells, which contributed to disease progression. With the advent of many biologicals targeting TNFalpha, IL-6, and IL-1 and their effective use in the treatment of autoimmune diseases, we propose that IL-7 and its receptor may be promising targets for biological agents in the treatment of autoimmunity.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-7/immunology , Lymphopenia/immunology , Models, Biological , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/transplantation , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL