Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Immunity ; 56(6): 1320-1340.e10, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37315535

ABSTRACT

CD8+ T cell exhaustion (Tex) limits disease control during chronic viral infections and cancer. Here, we investigated the epigenetic factors mediating major chromatin-remodeling events in Tex-cell development. A protein-domain-focused in vivo CRISPR screen identified distinct functions for two versions of the SWI/SNF chromatin-remodeling complex in Tex-cell differentiation. Depletion of the canonical SWI/SNF form, BAF, impaired initial CD8+ T cell responses in acute and chronic infection. In contrast, disruption of PBAF enhanced Tex-cell proliferation and survival. Mechanistically, PBAF regulated the epigenetic and transcriptional transition from TCF-1+ progenitor Tex cells to more differentiated TCF-1- Tex subsets. Whereas PBAF acted to preserve Tex progenitor biology, BAF was required to generate effector-like Tex cells, suggesting that the balance of these factors coordinates Tex-cell subset differentiation. Targeting PBAF improved tumor control both alone and in combination with anti-PD-L1 immunotherapy. Thus, PBAF may present a therapeutic target in cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Chromatin Assembly and Disassembly , Chromatin , Cell Differentiation , Epigenesis, Genetic
2.
Immunity ; 55(4): 671-685.e10, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417675

ABSTRACT

Interferon-gamma (IFN-γ) has pleiotropic effects on cancer immune checkpoint blockade (ICB), including roles in ICB resistance. We analyzed gene expression in ICB-sensitive versus ICB-resistant tumor cells and identified a strong association between interferon-mediated resistance and expression of Ripk1, a regulator of tumor necrosis factor (TNF) superfamily receptors. Genetic interaction screening revealed that in cancer cells, RIPK1 diverted TNF signaling through NF-κB and away from its role in cell death. This promoted an immunosuppressive chemokine program by cancer cells, enhanced cancer cell survival, and decreased infiltration of T and NK cells expressing TNF superfamily ligands. Deletion of RIPK1 in cancer cells compromised chemokine secretion, decreased ARG1+ suppressive myeloid cells linked to ICB failure in mice and humans, and improved ICB response driven by CASP8-killing and dependent on T and NK cells. RIPK1-mediated resistance required its ubiquitin scaffolding but not kinase function. Thus, cancer cells co-opt RIPK1 to promote cell-intrinsic and cell-extrinsic resistance to immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Interferons , Neoplasms , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Immunotherapy , Interferon-gamma/metabolism , Interferons/metabolism , Mice , NF-kappa B/metabolism , Neoplasms/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
Nat Immunol ; 14(6): 611-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644504

ABSTRACT

Natural T helper 17 (nTH17) cells are a population of interleukin 17 (IL-17)-producing cells that acquire effector function in the thymus during development. Here we demonstrate that the serine/threonine kinase Akt has a critical role in regulating nTH17 cell development. Although Akt and the downstream mTORC1-ARNT-HIFα axis were required for generation of inducible TH17 (iTH17) cells, nTH17 cells developed independently of mTORC1. In contrast, mTORC2 and inhibition of Foxo proteins were critical for development of nTH17 cells. Moreover, distinct isoforms of Akt controlled the generation of TH17 cell subsets, as deletion of Akt2, but not of Akt1, led to defective generation of iTH17 cells. These findings define mechanisms regulating nTH17 cell development and reveal previously unknown roles of Akt and mTOR in shaping subsets of T cells.


Subject(s)
Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Th17 Cells/immunology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/immunology , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoblotting , Interleukin-17/immunology , Interleukin-17/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 119(17): e2106083119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35446623

ABSTRACT

CD8 T cells mediate protection against intracellular pathogens and tumors. However, persistent antigen during chronic infections or cancer leads to T cell exhaustion, suboptimal functionality, and reduced protective capacity. Despite considerable work interrogating the transcriptional regulation of exhausted CD8 T cells (TEX), the posttranscriptional control of TEX remains poorly understood. Here, we interrogated the role of microRNAs (miRs) in CD8 T cells responding to acutely resolved or chronic viral infection and identified miR-29a as a key regulator of TEX. Enforced expression of miR-29a improved CD8 T cell responses during chronic viral infection and antagonized exhaustion. miR-29a inhibited exhaustion-driving transcriptional pathways, including inflammatory and T cell receptor signaling, and regulated ribosomal biogenesis. As a result, miR-29a fostered a memory-like CD8 T cell differentiation state during chronic infection. Thus, we identify miR-29a as a key regulator of TEX and define mechanisms by which miR-29a can divert exhaustion toward a more beneficial memory-like CD8 T cell differentiation state.


Subject(s)
MicroRNAs , Neoplasms , CD8-Positive T-Lymphocytes , Humans , Immunotherapy/methods , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/metabolism , Persistent Infection
5.
Nature ; 558(7709): 307-312, 2018 06.
Article in English | MEDLINE | ID: mdl-29849141

ABSTRACT

Cancer immunotherapy based on genetically redirecting T cells has been used successfully to treat B cell malignancies1-3. In this strategy, the T cell genome is modified by integration of viral vectors or transposons encoding chimaeric antigen receptors (CARs) that direct tumour cell killing. However, this approach is often limited by the extent of expansion and persistence of CAR T cells4,5. Here we report mechanistic insights from studies of a patient with chronic lymphocytic leukaemia treated with CAR T cells targeting the CD19 protein. Following infusion of CAR T cells, anti-tumour activity was evident in the peripheral blood, lymph nodes and bone marrow; this activity was accompanied by complete remission. Unexpectedly, at the peak of the response, 94% of CAR T cells originated from a single clone in which lentiviral vector-mediated insertion of the CAR transgene disrupted the methylcytosine dioxygenase TET2 gene. Further analysis revealed a hypomorphic mutation in this patient's second TET2 allele. TET2-disrupted CAR T cells exhibited an epigenetic profile consistent with altered T cell differentiation and, at the peak of expansion, displayed a central memory phenotype. Experimental knockdown of TET2 recapitulated the potency-enhancing effect of TET2 dysfunction in this patient's CAR T cells. These findings suggest that the progeny of a single CAR T cell induced leukaemia remission and that TET2 modification may be useful for improving immunotherapies.


Subject(s)
5-Methylcytosine/metabolism , Antigens, CD19/immunology , Dioxygenases/genetics , Immunotherapy/methods , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Adoptive Transfer , Aged , Alleles , Cell Differentiation , Clinical Trials as Topic , Clone Cells/cytology , Clone Cells/immunology , Dioxygenases/metabolism , Epigenesis, Genetic , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transgenes
6.
Crit Rev Immunol ; 40(2): 135-156, 2020.
Article in English | MEDLINE | ID: mdl-32749092

ABSTRACT

T lymphocytes undergo carefully orchestrated programming during development in the thymus and subsequently during differentiation in the periphery. This intricate specification allows for cell-type and context-specific transcriptional programs that regulate immune responses to infection and malignancy. Epigenetic changes, including histone modifications and covalent modification of DNA itself through DNA methylation, are now recognized to play a critical role in these cell-fate decisions. DNA methylation is mediated primarily by the actions of the DNA methyltransferase (DNMT) and ten-eleven-translocation (TET) families of epigenetic enzymes. In this review, we discuss the role of DNA methylation and its enzymatic regulators in directing the development and differentiation of CD4+ and CD8+ T-cells.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/immunology , DNA Methylation , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Animals , Biomarkers , Epigenesis, Genetic , Gene Expression Regulation , Humans , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/physiology
7.
Immunity ; 34(4): 492-504, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21497118

ABSTRACT

Polarized segregation of proteins in T cells is thought to play a role in diverse cellular functions including signal transduction, migration, and directed secretion of cytokines. Persistence of this polarization can result in asymmetric segregation of fate-determining proteins during cell division, which may enable a T cell to generate diverse progeny. Here, we provide evidence that a lineage-determining transcription factor, T-bet, underwent asymmetric organization in activated T cells preparing to divide and that it was unequally partitioned into the two daughter cells. This unequal acquisition of T-bet appeared to result from its asymmetric destruction during mitosis by virtue of concomitant asymmetric segregation of the proteasome. These results suggest a mechanism by which a cell may unequally localize cellular activities during division, thereby imparting disparity in the abundance of cell fate regulators in the daughter cells.


Subject(s)
Mitosis , Proteasome Endopeptidase Complex/metabolism , T-Box Domain Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Polarity , Cells, Cultured , Mice , Mice, Inbred C57BL , Phosphorylation , T-Box Domain Proteins/metabolism , T-Lymphocytes/enzymology
8.
J Immunol ; 200(1): 82-91, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29150566

ABSTRACT

T cell differentiation requires appropriate regulation of DNA methylation. In this article, we demonstrate that the methylcytosine dioxygenase ten-eleven translocation (TET)2 regulates CD8+ T cell differentiation. In a murine model of acute viral infection, TET2 loss promotes early acquisition of a memory CD8+ T cell fate in a cell-intrinsic manner without disrupting Ag-driven cell expansion or effector function. Upon secondary recall, TET2-deficient memory CD8+ T cells demonstrate superior pathogen control. Genome-wide methylation analysis identified a number of differentially methylated regions in TET2-deficient versus wild-type CD8+ T cells. These differentially methylated regions did not occur at the loci of differentially expressed memory markers; rather, several hypermethylated regions were identified in known transcriptional regulators of CD8+ T cell memory fate. Together, these data demonstrate that TET2 is an important regulator of CD8+ T cell fate decisions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Proto-Oncogene Proteins/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics
9.
Eur J Immunol ; 47(3): 516-526, 2017 03.
Article in English | MEDLINE | ID: mdl-28078715

ABSTRACT

Invariant NKT (iNKT) cells bridge innate and adaptive immunity by rapidly secreting cytokines and lysing targets following TCR recognition of lipid antigens. Based on their ability to secrete IFN-γ, IL-4 and IL-17A, iNKT-cells are classified as NKT-1, NKT-2, and NKT-17 subsets, respectively. The molecular pathways regulating iNKT-cell fate are not fully defined. Recent studies implicate Rictor, a required component of mTORC2, in the development of select iNKT-cell subsets, however these reports are conflicting. To resolve these questions, we used Rictorfl/fl CD4cre+ mice and found that Rictor is required for NKT-17 cell development and normal iNKT-cell cytolytic function. Conversely, Rictor is not absolutely required for IL-4 and IFN-γ production as peripheral iNKT-cells make copious amounts of these cytokines. Overall iNKT-cell numbers are dramatically reduced in the absence of Rictor. We provide data indicating Rictor regulates cell survival as well as proliferation of developing and mature iNKT-cells. Thus, mTORC2 regulates multiple aspects of iNKT-cell development and function.


Subject(s)
Carrier Proteins/metabolism , Multiprotein Complexes/metabolism , Natural Killer T-Cells/physiology , TOR Serine-Threonine Kinases/metabolism , Adaptive Immunity , Animals , Carrier Proteins/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Immunity, Innate , Lymphocyte Activation/genetics , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Proliferating Cell Nuclear Antigen/genetics , Radiation Chimera , Rapamycin-Insensitive Companion of mTOR Protein
10.
Immunity ; 28(3): 359-69, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18342008

ABSTRACT

The adaptor protein SLP76 directs signaling downstream of the T cell receptor (TCR) and is essential for thymocyte development. SLP76 contains three N-terminal tyrosines that are critical for its function. To define the role of these residues in thymocyte development, we generated two lines of "knock-in" mice, one expressing a mutation in tyrosine 145 (Y145F) and a second harboring two point mutations at tyrosines 112 and 128 (Y112-128F). We show here that although thymocyte development requires both Y145- and Y112-128-generated signals, selection was more dependent upon Y145. Although several proximal TCR signaling events were defective in both mutant mice, phosphorylation of the guanine nucleotide exchange factor, Vav1, and activation of Itk-dependent pathways were differentially affected by mutations at Y112-128 and Y145, respectively. Analysis of mice expressing one Y145F and one Y112-128F allele revealed that these mutants could complement one another in trans, demonstrating cooperativity between two or more SLP76 molecules. Thus, the N-terminal tyrosines of SLP76 are required for thymocyte selection but can function on separate molecules to support TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Differentiation/immunology , Phosphoproteins/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mice, Mutant Strains , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/immunology
11.
J Immunol ; 194(9): 4362-70, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25821220

ABSTRACT

Regulatory T cells (Tregs) are a subset of CD4(+) T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4(+) T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase-like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.


Subject(s)
Immunomodulation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Diglycerides/metabolism , Integrins/metabolism , Mice , Mice, Transgenic , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell/genetics
12.
J Immunol ; 193(2): 688-99, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24943215

ABSTRACT

T cell development in the thymus produces multiple lineages of cells, including innate T cells such as γδ TCR(+) cells, invariant NKT cells, mucosal-associated invariant T cells, and H2-M3-specific cells. Although innate cells are generally a minor subset of thymocytes, in several strains of mice harboring mutations in T cell signaling proteins or transcriptional regulators, conventional CD8(+) T cells develop as innate cells with characteristics of memory T cells. Thus, in Itk-deficient mice, mature CD4(-)CD8(+) (CD8 single-positive [SP]) thymocytes express high levels of the transcription factor eomesodermin (Eomes) and are dependent on IL-4 being produced in the thymic environment by a poorly characterized subset of CD4(+) thymocytes expressing the transcriptional regulator promyelocytic leukemia zinc finger. In this study, we show that a sizeable proportion of mature CD4(+)CD8(-) (CD4SP) thymocytes in itk(-/-) mice also develop as innate Eomes-expressing T cells. These cells are dependent on MHC class II and IL-4 signaling for their development, indicating that they are conventional CD4(+) T cells that have been converted to an innate phenotype. Surprisingly, neither CD4SP nor CD8SP innate Eomes(+) thymocytes in itk(-/-) or SLP-76(Y145F) mice are dependent on γδ T cells for their development. Instead, we find that the predominant population of Eomes(+) innate itk(-/-) CD4SP thymocytes is largely absent in mice lacking CD1d-specific invariant NKT cells, with no effect on innate itk(-/-) CD8SP thymocytes. In contrast, both subsets of innate Eomes(+)itk(-/-) T cells require the presence of a novel promyelocytic leukemia zinc finger-expressing, SLAM family receptor adapter protein-dependent thymocyte population that is essential for the conversion of conventional CD4(+) and CD8(+) T cells into innate T cells with a memory phenotype.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Protein-Tyrosine Kinases/immunology , Signal Transduction/immunology , Animals , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Flow Cytometry , Interleukin-15/deficiency , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-4/immunology , Interleukin-4/metabolism , Kruppel-Like Transcription Factors/immunology , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Rats , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Thymocytes/immunology , Thymocytes/metabolism
13.
Eur J Immunol ; 44(3): 785-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24307208

ABSTRACT

Autoreactive CD4(+) CD8(-) (CD4SP) thymocytes can be subjected to deletion when they encounter self-peptide during their development, but they can also undergo selection to become CD4SPFoxp3(+) Treg cells. We have analyzed the relationship between these distinct developmental fates using mice in which signals transmitted by the TCR have been attenuated by mutation of a critical tyrosine residue of the adapter protein SLP-76. In mice containing polyclonal TCR repertoires, the mutation caused increased frequencies of CD4SPFoxp3(+) thymocytes. CD4SP thymocytes expressing TCR Vß-chains that are subjected to deletion by endogenous retroviral superantigens were also present at increased frequencies, particularly among Foxp3(+) thymocytes. In transgenic mice in which CD4SP thymocytes expressing an autoreactive TCR undergo both deletion and Treg-cell formation in response to a defined self-peptide, SLP-76 mutation abrogated deletion of autoreactive CD4SP thymocytes. Notably, Foxp3(+) Treg-cell formation still occurred, albeit with a reduced efficiency, and the mutation was also associated with decreased Nur77 expression by the autoreactive CD4SP thymocytes. These studies provide evidence that the strength of the TCR signal can play a direct role in directing the extent of both thymocyte deletion and Treg-cell differentiation, and suggest that distinct TCR signaling thresholds and/or pathways can promote CD4SP thymocyte deletion versus Treg-cell formation.


Subject(s)
Autoantigens/immunology , Clonal Deletion/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigen Presentation , Autoantigens/chemistry , Autoimmunity , Clonal Deletion/genetics , Forkhead Transcription Factors/metabolism , Gene Expression , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation , Peptides/chemistry , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction
14.
J Immunol ; 191(1): 228-37, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23709682

ABSTRACT

Clinical evidence for a more active immune response in humans compared with our closest hominid relative, the chimpanzee, includes the progression of HIV infection to AIDS, hepatitis B- and C-related inflammation, autoimmunity, and unwanted harmful immune responses to viral gene transfer vectors. Humans have a unique mutation of the enzyme CMP-N-acetylneuraminic acid hydroxylase (CMAH), causing loss of expression of the sialic acid Neu5Gc. This mutation, occurring 2 million years ago, likely altered the expression and function of ITIM-bearing inhibitory receptors (Siglecs) that bind sialic acids. Previous work showed that human T cells proliferate faster than chimpanzee T cells upon equivalent stimulation. In this article, we report that Cmah(-/-) mouse T cells proliferate faster and have greater expression of activation markers than wild-type mouse T cells. Metabolically reintroducing Neu5Gc diminishes the proliferation and activation of both human and murine Cmah(-/-) T cells. Importantly, Cmah(-/-) mice mount greater T cell responses to an adenovirus encoding an adeno-associated virus capsid transgene. Upon lymphocytic choriomeningitis virus infection, Cmah(-/-) mice make more lymphocytic choriomeningitis virus-specific T cells than WT mice, and these T cells are more polyfunctional. Therefore, a uniquely human glycosylation mutation, modeled in mice, leads to a more proliferative and active T cell population. These findings in a human-like mouse model have implications for understanding the hyperimmune responses that characterize some human diseases.


Subject(s)
Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Dependovirus/genetics , Dependovirus/immunology , Dependovirus/metabolism , Glycosylation , Humans , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , T-Lymphocyte Subsets/enzymology , Up-Regulation/genetics , Up-Regulation/immunology
15.
Cell Mol Life Sci ; 70(13): 2271-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23052209

ABSTRACT

Interleukin (IL)-17 is a pro-inflammatory cytokine that plays critical roles in host defense against extracellular bacteria and fungi and also in the pathogenesis of autoimmune diseases. While CD4+ TCRαß+ T helper (Th) 17 cells are the best-described cellular source of IL-17, many innate-like T cells are in fact potent producers of IL-17. Given the increasing interest in therapeutic modulation of the IL-17 axis, it is crucial to better understand the cellular origins of IL-17 in various infection and diseases settings. While the diverse population of IL-17-producing T cells share many common characteristics, notable differences also exist. In this review, we discuss the heterogeneity of IL-17-producing T cell types focusing on their development, regulation, and function.


Subject(s)
Th17 Cells/classification , Animals , Autoimmunity , Cytokines/metabolism , Humans , Models, Immunological , Neoplasms/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Th17 Cells/cytology , Th17 Cells/physiology , Transcription Factors/metabolism , Transcription Factors/physiology
16.
bioRxiv ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38915532

ABSTRACT

Akt1 and Akt2, isoforms of the serine threonine kinase Akt, are essential for T cell development. However, their role in peripheral T cell differentiation remains undefined. Using mice with germline deletions of either Akt1 or Akt2, we found that both isoforms are important for Th17 differentiation, although Akt2 loss had a greater impact than loss of Akt1. In contrast to defective IL-17 production, Akt2 -/- T cells exhibited enhanced IL-4 production in vitro under Th2 polarizing conditions. In vivo , Akt2 -/- mice displayed significantly diminished IL-17A and GM-CSF production following immunization with myelin oligodendrocyte glycoprotein (MOG). This dampened response was associated with further alterations in Th cell differentiation including decreased IFNγ production but preserved IL-4 production, and preferential expansion of regulatory T cells compared to non-regulatory CD4 T cells. Taken together, we identify Akt2 as an important signaling molecule in regulating peripheral CD4 T cell responses.

17.
J Immunol ; 187(9): 4459-66, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21949020

ABSTRACT

The adapter protein Src homology 2 (SH2) domain-containing leukocyte protein of 76 kDa (SLP-76) is critical for multiple aspects of T cell development and function. Through its protein-binding domains, SLP-76 serves as a platform for the assembly of multiple enzymes and adapter proteins that function together to activate second messengers required for TCR signal propagation. The N terminus of SLP-76, which contains three tyrosines that serve as docking sites for SH2 domain-containing proteins, and the central proline-rich region of SLP-76 have been well studied and are known to be important for both thymocyte selection and activation of peripheral T cells. Less is known about the function of the C-terminal SH2 domain of SLP-76. This region inducibly associates with ADAP and HPK1. Combining regulated deletion of endogenous SLP-76 with transgenic expression of a SLP-76 SH2 domain mutant, we demonstrate that the SLP-76 SH2 domain is required for peripheral T cell activation and positive selection of thymocytes, a function not previously attributed to this region. This domain is also important for T cell proliferation, IL-2 production, and phosphorylation of protein kinase D and IκB. ADAP-deficient T cells display similar, but in some cases less severe, defects despite phosphorylation of a negative regulatory site on SLP-76 by HPK1, a function that is lost in SLP-76 SH2 domain mutant T cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Phosphoproteins/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , src Homology Domains/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation/genetics , Humans , Jurkat Cells , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphoproteins/deficiency , Phosphoproteins/genetics , Signal Transduction/genetics , Signal Transduction/immunology , src Homology Domains/genetics
18.
J Immunol ; 186(8): 4573-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21383242

ABSTRACT

Conventional and nonconventional T cell development occur in the thymus. Nonconventional thymocytes that bear characteristics typically associated with innate immune cells are termed innate-like lymphocytes (ILLs). Mice harboring a tyrosine to phenylalanine mutation in the adaptor protein Src homology 2 domain-containing leukocyte protein of 76 kDa at residue 145 (Y145F mice) develop an expanded population of CD8(+)CD122(+)CD44(+) ILLs, typified by expression of the T-box transcription factor eomesodermin. Y145F mice also have an expanded population of γδ T cells that produce copious amounts of IL-4 via a mechanism that is dependent on the BTB-ZF transcription factor promyelocytic leukemia zinc finger. Using mice with T cell-specific deletion of Eomes, we demonstrate that this transcription factor is required for CD8(+) ILL development in Y145F as well as wild-type mice. Moreover, we show that promyelocytic leukemia zinc finger and IL-4 are also required for the generation of this ILL population. Taken together, these data shed light on the cell-intrinsic and cell-extrinsic factors that drive CD8(+) ILL differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD8-Positive T-Lymphocytes/immunology , Kruppel-Like Transcription Factors/immunology , Phosphoproteins/immunology , T-Box Domain Proteins/immunology , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phosphoproteins/genetics , Promyelocytic Leukemia Zinc Finger Protein , T-Box Domain Proteins/genetics , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Zinc Fingers/genetics , Zinc Fingers/immunology
19.
bioRxiv ; 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36824931

ABSTRACT

T cell exhaustion (T EX ) impairs the ability of T cells to clear chronic infection or cancer. While exhausted T cells are hypofunctional, some exhausted T cells retain effector gene signatures, a feature that is associated with expression of KLRs (killer lectin-like receptors). Although KLR + T cells may improve control of chronic antigen, the signaling molecules regulating this population are poorly understood. Using scRNA-seq, flow cytometry, RNA velocity, and scTCR-seq, we demonstrate that deleting the pseudokinase Trib1 shifts T EX towards CX3CR1 + intermediates (T INT ) with robust enrichment of KLR + CD8 + T cells (T KLR ) via clonal T cell expansion. These changes are associated with globally increased KLR gene expression throughout the exhaustion program. Further, Trib1 loss augments anti-PD-L1 blockade to improve viral clearance by expanding the T KLR population. Together, these data identify Trib1 as an important regulator of T cell exhaustion whose targeting enhances the KLR + effector state and improves the response to checkpoint inhibitor therapy.

20.
Cell Rep ; 42(8): 112905, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37527035

ABSTRACT

CD8+ T cell exhaustion (TEX) impairs the ability of T cells to clear chronic infection or cancer. While TEX are hypofunctional, some TEX retain effector gene signatures, a feature associated with killer lectin-like receptor (KLR) expression. Although KLR+ TEX (TKLR) may improve control of chronic antigen, the signaling molecules regulating this population are poorly understood. Using single-cell RNA sequencing (scRNA-seq), flow cytometry, RNA velocity, and single-cell T cell receptor sequencing (scTCR-seq), we demonstrate that deleting the pseudokinase Trib1 shifts TEX toward CX3CR1+ intermediates with robust enrichment of TKLR via clonal T cell expansion. Adoptive transfer studies demonstrate this shift toward CD8+ TKLR in Trib1-deficient cells is CD8 intrinsic, while CD4-depletion studies demonstrate CD4+ T cells are required for improved viral control in Trib1 conditional knockout mice. Further, Trib1 loss augments anti-programmed death-ligand 1 (PD-L1) blockade to improve viral clearance. These data identify Trib1 as an important regulator of CD8+ TEX whose targeting enhances the TKLR effector state and improves checkpoint inhibitor therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Mice , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL