Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 24(6): 1036-1048, 2023 06.
Article in English | MEDLINE | ID: mdl-37106040

ABSTRACT

Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.


Subject(s)
Hypersensitivity , Janus Kinase Inhibitors , Humans , Interleukin-9/genetics , T-Lymphocytes, Helper-Inducer , STAT5 Transcription Factor/genetics , Chromatin/genetics , Inflammation , Hypersensitivity/genetics , Cell Differentiation , STAT6 Transcription Factor
2.
Nat Immunol ; 19(9): 986-1000, 2018 09.
Article in English | MEDLINE | ID: mdl-30127432

ABSTRACT

Gain-of-function mutations in the gene encoding the phosphatidylinositol-3-OH kinase catalytic subunit p110δ (PI3Kδ) result in a human primary immunodeficiency characterized by lymphoproliferation, respiratory infections and inefficient responses to vaccines. However, what promotes these immunological disturbances at the cellular and molecular level remains unknown. We generated a mouse model that recapitulated major features of this disease and used this model and patient samples to probe how hyperactive PI3Kδ fosters aberrant humoral immunity. We found that mutant PI3Kδ led to co-stimulatory receptor ICOS-independent increases in the abundance of follicular helper T cells (TFH cells) and germinal-center (GC) B cells, disorganized GCs and poor class-switched antigen-specific responses to immunization, associated with altered regulation of the transcription factor FOXO1 and pro-apoptotic and anti-apoptotic members of the BCL-2 family. Notably, aberrant responses were accompanied by increased reactivity to gut bacteria and a broad increase in autoantibodies that were dependent on stimulation by commensal microbes. Our findings suggest that proper regulation of PI3Kδ is critical for ensuring optimal host-protective humoral immunity despite tonic stimulation from the commensal microbiome.


Subject(s)
B-Lymphocytes/physiology , Gastrointestinal Microbiome/immunology , Germinal Center/physiology , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes, Helper-Inducer/physiology , Animals , Autoantibodies/blood , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Immunity, Humoral/genetics , Immunoglobulin Class Switching/genetics , Immunologic Deficiency Syndromes/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
Immunol Rev ; 291(1): 154-173, 2019 09.
Article in English | MEDLINE | ID: mdl-31402502

ABSTRACT

Phosphatidylinositol 3 kinases (PI3K) are a family of lipid kinases that are activated by a variety of cell-surface receptors, and regulate a wide range of downstream readouts affecting cellular metabolism, growth, survival, differentiation, adhesion, and migration. The importance of these lipid kinases in lymphocyte signaling has recently been highlighted by genetic analyses, including the recognition that both activating and inactivating mutations of the catalytic subunit of PI3Kδ, p110δ, lead to human primary immunodeficiencies. In this article, we discuss how studies on the human genetic disorder "Activated PI3K-delta syndrome" and mouse models of this disease (Pik3cdE1020K/+ mice) have provided fundamental insight into pathways regulated by PI3Kδ in T and B cells and their contribution to lymphocyte function and disease, including responses to commensal bacteria and the development of autoimmunity and tumors. We highlight critical roles of PI3Kδ in T follicular helper cells and the orchestration of the germinal center reaction, as well as in CD8+ T-cell function. We further  present data demonstrating the ability of the AKT-resistant FOXO1AAA mutant to rescue IgG1 class switching defects in Pik3cdE1020K/+ B cells, as well as data supporting a role for PI3Kδ in promoting multiple T-helper effector cell lineages.


Subject(s)
B-Lymphocytes/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , Autoimmunity , B-Lymphocytes/immunology , Biomarkers , Disease Susceptibility , Energy Metabolism , Humans , Immunotherapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/therapy , Primary Immunodeficiency Diseases/etiology , Primary Immunodeficiency Diseases/metabolism , T-Lymphocytes/immunology
5.
Hum Mol Genet ; 28(17): 2920-2936, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31194862

ABSTRACT

Proteus syndrome is a mosaic, progressive overgrowth disorder caused by a somatic activating variant c.49G > A p.(E17K) in AKT1. The presentation in affected individuals is variable, with a diversity of tissues demonstrating abnormalities. Common manifestations include skin and bony overgrowth, vascular malformations (VMs), cysts and benign tumors. We used two methods to create mouse models that had endogenously-regulated mosaic expression of the Proteus syndrome variant. Variant allele fractions (VAFs) ranged from 0% to 50% across numerous tissues in 44 Proteus syndrome mice. Mice were phenotypically heterogeneous with lesions rarely observed before 12 months of age. VMs were the most frequent finding with a total of 69 found in 29 of 44 Proteus syndrome mice. Twenty-eight cysts and ectasia, frequently biliary, were seen in 22 of 44 Proteus syndrome mice. Varying levels of mammary hyperplasia were seen in 10 of 16 female Proteus syndrome mice with other localized regions of hyperplasia and stromal expansion noted in several additional animals. Interestingly, 27 of 31 Proteus syndrome animals had non-zero blood VAF that is in contrast to the human disorder where it is rarely seen in peripheral blood. Identification of variant-positive cells by green fluorescent protein (GFP) staining in chimeric Proteus syndrome mice showed that in some lesions, hyperplastic cells were predominantly GFP/Akt1E17K-positive and showed increased pAKT signal compared to GFP-negative cells. However, hyperplastic mammary epithelium was a mixture of GFP/Akt1E17K-positive and negative cells with some GFP/Akt1E17K-negative cells also having increased pAKT signal suggesting that the variant-positive cells can induce lesion formation in a non-cell autonomous manner.


Subject(s)
Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Phenotype , Proteus Syndrome/genetics , Alleles , Animals , Biopsy , Genetic Association Studies/methods , Genetic Loci , Genotype , Humans , Mice , Proteus Syndrome/diagnosis , Proto-Oncogene Proteins c-akt/genetics
6.
Immunity ; 32(2): 253-65, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20153220

ABSTRACT

CD4(+) T cells deficient in signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) exhibit a selective impairment in adhesion to antigen-presenting B cells but not dendritic cells (DCs), resulting in defective germinal center formation. However, the nature of this selective adhesion defect remained unclear. We found that whereas T cell:DC interactions were primarily integrin dependent, T cell:B cell interactions had both an early integrin-dependent phase and a sustained phase that also required SAP. We further found that the SLAM family member CD84 was required for prolonged T cell:B cell contact, optimal T follicular helper function, and germinal center formation in vivo. Moreover, both CD84 and another SLAM member, Ly108, mediated T cell adhesion and participated in stable T cell:B cell interactions in vitro. Our results reveal insight into the dynamic regulation of T cell:B cell interactions and identify SLAM family members as critical components of sustained T cell:B cell adhesion required for productive humoral immunity.


Subject(s)
B-Lymphocytes/metabolism , Cell Adhesion/immunology , Germinal Center/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Germinal Center/pathology , Interferon-gamma/metabolism , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocytes/immunology , T-Lymphocytes/pathology
7.
Immunity ; 31(4): 587-97, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818650

ABSTRACT

T helper 17 (Th17) cells play major roles in autoimmunity and bacterial infections, yet how T cell receptor (TCR) signaling affects Th17 cell differentiation is relatively unknown. We demonstrate that CD4(+) T cells lacking Itk, a tyrosine kinase required for full TCR-induced phospholipase C-gamma (PLC-gamma1) activation, exhibit decreased interleukin-17A (IL-17A) expression in vitro and in vivo, despite relatively normal expression of retinoic acid receptor-related orphan receptor-gammaT (ROR-gammaT) and IL-17F. IL-17A expression was rescued by pharmacologically induced Ca(2+) influx or constitutively activated nuclear factor of activated T cells (NFAT). Conversely, decreased TCR stimulation or calcineurin inhibition preferentially reduced IL-17A expression. We further found that the promoter of Il17a but not Il17f has a conserved NFAT binding site that bound NFATc1 in wild-type but not Itk-deficient cells, even though both exhibited open chromatin conformations. Finally, Itk(-/-) mice also showed differential regulation of IL-17A and IL-17F in vivo. Our results suggest that Itk specifically couples TCR signaling to Il17a expression and the differential regulation of Th17 cell cytokines through NFATc1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Interleukin-17/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cytokines/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Promoter Regions, Genetic/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , Signal Transduction/immunology
8.
J Immunol ; 194(8): 3567-82, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25786692

ABSTRACT

The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Interleukin-9/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Asthma/genetics , Asthma/pathology , Cell Differentiation/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-9/genetics , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
9.
Cereb Cortex ; 25(10): 3572-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25209608

ABSTRACT

Although long noncoding RNAs (lncRNAs) are proposed to play essential roles in mammalian neurodevelopment, we know little of their functions from their disruption in vivo. Combining evidence for evolutionary constraint and conserved expression data, we previously identified candidate lncRNAs that might play important and conserved roles in brain function. Here, we demonstrate that the sequence and neuronal transcription of lncRNAs transcribed from the previously uncharacterized Visc locus are conserved across diverse mammals. Consequently, one of these lncRNAs, Visc-2, was selected for targeted deletion in the mouse, and knockout animals were subjected to an extremely detailed anatomical and behavioral characterization. Despite a neurodevelopmental expression pattern of Visc-2 that is highly localized to the cortex and sites of neurogenesis, anomalies in neither cytoarchitecture nor neuroproliferation were identified in knockout mice. In addition, no abnormal motor, sensory, anxiety, or cognitive behavioral phenotypes were observed. These results are important because they contribute to a growing body of evidence that lncRNA loci contribute on average far less to brain and biological functions than protein-coding loci. A high-throughput knockout program focussing on lncRNAs, similar to that currently underway for protein-coding genes, will be required to establish the distribution of their organismal functions.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Conserved Sequence/genetics , RNA, Long Noncoding/genetics , Animals , Anxiety/genetics , Base Sequence/genetics , Brain/cytology , Brain/growth & development , Evolution, Molecular , Female , Male , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Phenotype , RNA, Long Noncoding/metabolism
10.
J Immunol ; 190(5): 2121-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355739

ABSTRACT

The promyelocytic zinc finger transcription factor (PLZF) is required for the development of activated phenotypes in NKT and other innate T lymphocytes. Although strong TCR stimulation has been implicated in the induction of PLZF, the factors regulating PLZF expression are incompletely understood. We show in this study that costimulation of preselection double-positive thymocytes through the signaling lymphocyte activation molecule family receptor Ly108 markedly enhanced PLZF expression compared with that induced by TCR stimulation alone. Costimulation with Ly108 increased expression of early growth response protein (Egr)-2 and binding of Egr-2 to the promoter of Zbtb16, which encodes PLZF, and resulted in PLZF levels similar to those seen in NKT cells. In contrast, costimulation with anti-CD28 failed to enhance Egr-2 binding and Zbtb16 expression. Moreover, mice lacking Ly108 showed decreased numbers of PLZF-expressing CD4(+) T cells. Together, these results support a potential role for Ly108 in the induction of PLZF.


Subject(s)
Antigens, Ly/genetics , Cell Differentiation/immunology , Kruppel-Like Transcription Factors/genetics , Thymocytes/cytology , Animals , Antibodies/pharmacology , Antigens, Ly/immunology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Gene Expression Regulation/drug effects , Kruppel-Like Transcription Factors/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Thymocytes/drug effects , Thymocytes/immunology
11.
J Exp Med ; 221(5)2024 May 06.
Article in English | MEDLINE | ID: mdl-38557723

ABSTRACT

CD4+ T cells are vital for host defense and immune regulation. However, the fundamental role of CD4 itself remains enigmatic. We report seven patients aged 5-61 years from five families of four ancestries with autosomal recessive CD4 deficiency and a range of infections, including recalcitrant warts and Whipple's disease. All patients are homozygous for rare deleterious CD4 variants impacting expression of the canonical CD4 isoform. A shorter expressed isoform that interacts with LCK, but not HLA class II, is affected by only one variant. All patients lack CD4+ T cells and have increased numbers of TCRαß+CD4-CD8- T cells, which phenotypically and transcriptionally resemble conventional Th cells. Finally, patient CD4-CD8- αß T cells exhibit intact responses to HLA class II-restricted antigens and promote B cell differentiation in vitro. Thus, compensatory development of Th cells enables patients with inherited CD4 deficiency to acquire effective cellular and humoral immunity against an unexpectedly large range of pathogens. Nevertheless, CD4 is indispensable for protective immunity against at least human papillomaviruses and Trophyrema whipplei.


Subject(s)
CD4-Positive T-Lymphocytes , T-Lymphocytes, Helper-Inducer , Humans , CD8-Positive T-Lymphocytes , Lymphocyte Activation , HLA Antigens , Protein Isoforms/metabolism
12.
Proc Natl Acad Sci U S A ; 107(45): 19455-60, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974963

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is a type I cytokine that plays essential roles in allergic/inflammatory skin and airway disorders, in helminth infections, and in regulating intestinal immunity. TSLP signals via IL-7Rα and a specific TSLPR subunit that is highly related to the common cytokine receptor γ chain, γ(c). Although TSLP has effects on a broad range of hematopoetic cells and can induce STAT5 phosphorylation, TSLP was reported to not signal via JAK kinases, and the mechanism by which TSLP regulates STAT5 phosphorylation has been unclear. We now demonstrate the role of JAK1 and JAK2 in TSLP-mediated STAT5 phosphorylation in mouse and human primary CD4(+) T cells, in contrast to the known activation of JAK1 and JAK3 by the related cytokine, IL-7. We also show that just as JAK1 interacts with IL-7Rα, JAK2 is associated with TSLPR protein. Moreover, we demonstrate the importance of STAT5 activation for TSLP-mediated survival and proliferation of CD4(+) T cells. These findings clarify the basis for TSLP-mediated signaling and provide an example wherein a cytokine uses JAK1 and JAK2 to mediate the activation of STAT5.


Subject(s)
Cytokines/metabolism , Interleukin-7/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Humans , Mice , Phosphorylation , Signal Transduction , Thymic Stromal Lymphopoietin
13.
Res Sq ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36789409

ABSTRACT

Background: Modulation of metabolic flux through pyruvate dehydrogenase complex (PDC) plays an important role in T cell activation and differentiation. PDC sits at the transition between glycolysis and the tricarboxylic acid cycle and is a major producer of acetyl-CoA, marking it as a potential metabolic and epigenetic node. Methods: To understand the role of pyruvate dehydrogenase complex in T cell differentiation, we generated mice deficient in T cell pyruvate dehydrogenase E1A (Pdha) subunit using a CD4-cre recombinase-based strategy. To control for the contribution of exogenous metabolites in vivo, we conducted our T cell functional studies in vitro. T cells were differentiated into memory and effector T cells using standardized protocols. Cells were analyzed using stable isotopic tracing studies, metabolomics, RNAseq, ATACseq, ChIPseq and histone proteomics. Results: Herein, we show that genetic ablation of PDC activity in T cells (TPdh-/-) leads to marked perturbations in glycolysis, the tricarboxylic acid cycle, and OXPHOS. Due to depressed OXPHOS, TPdh-/-T cells became dependent upon substrate level phosphorylation via glycolysis. Due to the block of PDC activity, histone acetylation was reduced, as were most other types of post translational modifications. Transcriptional and functional profiling revealed abnormal CD8+ memory T cell differentiation in vitro. Conclusions: Collectively, our data indicate that PDC integrates the metabolome and epigenome in memory T cell differentiation. Targeting this metabolic and epigenetic node can have widespread ramifications on cellular function.

14.
Res Sq ; 2023 May 10.
Article in English | MEDLINE | ID: mdl-37215014

ABSTRACT

Modulation of metabolic flux through pyruvate dehydrogenase complex (PDC) plays an important role in T cell activation and differentiation. PDC sits at the transition between glycolysis and the tricarboxylic acid cycle and is a major producer of acetyl-CoA, marking it as a potential metabolic and epigenetic node To understand the role of pyruvate dehydrogenase complex in T cell differentiation, we generated mice deficient in T cell pyruvate dehydrogenase E1A (Pdha) subunit using a CD4-cre recombinase-based strategy. Herein, we show that genetic ablation of PDC activity in T cells (TPdh-/-) leads to marked perturbations in glycolysis, the tricarboxylic acid cycle, and OXPHOS. TPdh-/- T cells became dependent upon substrate level phosphorylation via glycolysis, secondary to depressed OXPHOS. Due to the block of PDC activity, histone acetylation was also reduced, including H3K27, a critical site for CD8+ TM differentiation. Transcriptional and functional profiling revealed abnormal CD8+ TM differentiation in vitro. Collectively, our data indicate that PDC integrates the metabolome and epigenome in CD8+ memory T cell differentiation. Targeting this metabolic and epigenetic node can have widespread ramifications on cellular function.

15.
J Immunol ; 185(5): 2819-27, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20668219

ABSTRACT

Signaling lymphocytic activation molecule-associated protein (SAP), an adaptor molecule that recruits Fyn to the signaling lymphocytic activation molecule (SLAM) family of immunomodulatory receptors, is mutated in X-linked lymphoproliferative disease. CD4(+) T cells from SAP-deficient mice have defective TCR-induced and follicular Th cell IL-4 production and impaired T cell-mediated help for germinal center formation; however, the downstream intermediates contributing to these defects remain unclear. We previously found that SAP-deficient CD4(+) T cells exhibit decreased protein kinase C (PKC)-theta recruitment upon TCR stimulation. We demonstrate in this paper using GST pulldowns and coimmunoprecipitation studies that SAP constitutively associates with PKC- in T cells. SAP-PKC-theta interactions required R78 of SAP, a residue previously implicated in Fyn recruitment, yet SAP's interactions with PKC-theta occurred independent of phosphotyrosine binding and Fyn. Overexpression of SAP in T cells increased and sustained PKC-theta recruitment to the immune synapse and elevated IL-4 production in response to TCR plus SLAM-mediated stimulation. Moreover, PKC-theta, like SAP, was required for SLAM-mediated increases in IL-4 production, and, conversely, membrane-targeted PKC-theta mutants rescued IL-4 expression in SAP(-/-) CD4(+) T cells, providing genetic evidence that PKC-theta is a critical component of SLAM/SAP-mediated pathways that influence TCR-driven IL-4 production.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Interleukin-4/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Gene Expression Regulation/immunology , Humans , Interleukin-4/deficiency , Interleukin-4/genetics , Isoenzymes/deficiency , Jurkat Cells , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Kinase C/deficiency , Protein Kinase C-theta , Protein Transport/genetics , Protein Transport/immunology , Receptors, Cell Surface/deficiency , Signal Transduction/genetics , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Up-Regulation/genetics , Up-Regulation/immunology
17.
Nat Commun ; 13(1): 805, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145086

ABSTRACT

T follicular helper (Tfh) cells provide signals to initiate and maintain the germinal center (GC) reaction and are crucial for the generation of robust, long-lived antibody responses, but how the GC microenvironment affects Tfh cells is not well understood. Here we develop an in vivo T cell-intrinsic CRISPR-knockout screen to evaluate Tfh and Th1 cells in an acute viral infection model to identify regulators of Tfh cells in their physiological setting. Using a screen of druggable-targets, alongside genetic, transcriptomic and cellular analyses, we identify a function of HIF-1α in suppressing mTORC1-mediated and Myc-related pathways, and provide evidence that VHL-mediated degradation of HIF-1α is required for Tfh development; an expanded in vivo CRISPR screen reveals multiple components of these pathways that regulate Tfh versus Th1 cells, including signaling molecules, cell-cycle regulators, nutrient transporters, metabolic enzymes and autophagy mediators. Collectively, our data serve as a resource for studying Tfh versus Th1 decisions, and implicate the VHL-HIF-1α axis in fine-tuning Tfh generation.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Antibody Formation , Cell Differentiation/immunology , Gene Expression , Gene Knockout Techniques , Germinal Center/immunology , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Humoral/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Virus Diseases/immunology
18.
Cell Rep ; 37(2): 109804, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644563

ABSTRACT

Patients with activated phosphatidylinositol 3-kinase delta (PI3Kδ) syndrome (APDS) present with sinopulmonary infections, lymphadenopathy, and cytomegalvirus (CMV) and/or Epstein-Barr virus (EBV) viremia, yet why patients fail to clear certain chronic viral infections remains incompletely understood. Using patient samples and a mouse model (Pik3cdE1020K/+ mice), we demonstrate that, upon activation, Pik3cdE1020K/+ CD8+ T cells exhibit exaggerated features of effector populations both in vitro and after viral infection that are associated with increased Fas-mediated apoptosis due to sustained FoxO1 phosphorylation and Fasl derepression, enhanced mTORC1 and c-Myc signatures, metabolic perturbations, and an altered chromatin landscape. Conversely, Pik3cdE1020K/+ CD8+ cells fail to sustain expression of proteins critical for central memory, including TCF1. Strikingly, activated Pik3cdE1020K/+ CD8+ cells exhibit altered transcriptional and epigenetic circuits characterized by pronounced interleukin-2 (IL-2)/STAT5 signatures and heightened IL-2 responses that prevent differentiation to memory-like cells in IL-15. Our data position PI3Kδ as integrating multiple signaling nodes that promote CD8+ T cell effector differentiation, providing insight into phenotypes of patients with APDS.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Chromatin Assembly and Disassembly , Chromatin/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Immunologic Memory , Primary Immunodeficiency Diseases/enzymology , Transcription, Genetic , Virus Diseases/enzymology , Adolescent , Adult , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Case-Control Studies , Child , Chromatin/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/immunology , Disease Models, Animal , Enzyme Activation , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Signal Transduction , Virus Diseases/genetics , Virus Diseases/immunology
19.
J Immunol ; 181(9): 6125-31, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941202

ABSTRACT

Itk and Txk/Rlk are Tec family kinases expressed in T cells. Itk is expressed in both Th1 and Th2 cells. By contrast, Txk is preferentially expressed in Th1 cells. Although Itk is required for Th2 responses in vivo and Txk is suggested to regulate IFN-gamma expression and Th1 responses, it remains unclear whether these kinases have distinct roles in Th cell differentiation/function. We demonstrate here that Txk-null CD4(+) T cells are capable of producing both Th1 and Th2 cytokines similar to those produced by wild-type CD4(+) T cells. To further examine whether Itk and Txk play distinct roles in Th cell differentiation and function, we examined Itk-null mice carrying a transgene that expresses Txk at levels similar to the expression of Itk in Th2 cells. Using two Th2 model systems, allergic asthma and schistosome egg-induced lung granulomas, we found that the Txk transgene rescued Th2 cytokine production and all Th2 symptoms without notable enhancement of IFN-gamma expression. These results suggest that Txk is not a specific regulator of Th1 responses. Importantly, they suggest that Itk and Txk exert their effects on Th cell differentiation/function at the level of expression.


Subject(s)
Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Th1 Cells/enzymology , Th1 Cells/immunology , Th2 Cells/enzymology , Th2 Cells/immunology , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/enzymology , Asthma/immunology , Asthma/parasitology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Schistosomiasis mansoni/enzymology , Schistosomiasis mansoni/immunology , Th1 Cells/metabolism , Th1 Cells/parasitology , Th2 Cells/metabolism , Th2 Cells/parasitology
20.
Nucleic Acids Res ; 36(18): e117, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710883

ABSTRACT

We evaluate here the use of real-time quantitative PCR (q-PCR) as a method for screening for homologous recombinants generated in mammalian cells from either conventional gene-targeting constructs or whole BAC-based constructs. Using gene-targeted events at different loci, we show that q-PCR is a highly sensitive and accurate method for screening for conventional gene targeting that can reduce the number of clones requiring follow-up screening by Southern blotting. We further compared q-PCR to fluorescent in situ hybridization (FISH) for the detection of gene-targeting events using full-length BAC-based constructs designed to introduce mutations either into one gene or simultaneously into two adjacent genes. We find that although BAC-based constructs appeared to have high rates of homologous recombination when evaluated by FISH, screening by FISH was prone to false positives that were detected by q-PCR. Our results demonstrate the utility of q-PCR as a screening tool for gene targeting and further highlight potential problems with the use of whole BAC-based constructs for homologous recombination.


Subject(s)
Chromosomes, Artificial, Bacterial , Gene Targeting/methods , Polymerase Chain Reaction/methods , Recombination, Genetic , Animals , Cell Line , In Situ Hybridization, Fluorescence , Mice
SELECTION OF CITATIONS
SEARCH DETAIL