Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Annu Rev Immunol ; 38: 397-419, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31990620

ABSTRACT

T cell development involves stepwise progression through defined stages that give rise to multiple T cell subtypes, and this is accompanied by the establishment of stage-specific gene expression. Changes in chromatin accessibility and chromatin modifications accompany changes in gene expression during T cell development. Chromatin-modifying enzymes that add or reverse covalent modifications to DNA and histones have a critical role in the dynamic regulation of gene expression throughout T cell development. As each chromatin-modifying enzyme has multiple family members that are typically all coexpressed during T cell development, their function is sometimes revealed only when two related enzymes are concurrently deleted. This work has also revealed that the biological effects of these enzymes often involve regulation of a limited set of targets. The growing diversity in the types and sites of modification, as well as the potential for a single enzyme to catalyze multiple modifications, is also highlighted.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Lymphopoiesis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Acetylation , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Histones , Humans , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Methylation , Protein Processing, Post-Translational , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Ubiquitination
2.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38677292

ABSTRACT

Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Glycolysis , HMGB Proteins , Immunity, Innate , Lymphocytes , Mice, Knockout , Animals , Mice , Adaptation, Physiological/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Hexokinase/metabolism , Hexokinase/genetics , Interleukin-17/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Trans-Activators/metabolism , Trans-Activators/genetics , HMGB Proteins/genetics , HMGB Proteins/immunology , HMGB Proteins/metabolism
3.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33951416

ABSTRACT

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Subject(s)
COVID-19/immunology , COVID-19/virology , Cell Self Renewal/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , SARS-CoV-2/immunology , Biomarkers , COVID-19/metabolism , Cytokines/metabolism , Disease Susceptibility/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Signal Transduction
4.
Immunity ; 50(5): 1188-1201.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31053504

ABSTRACT

Lymph nodes (LNs) play critical roles in adaptive immunity by concentrating in one location the antigens, antigen-presenting cells, and antigen-responsive lymphocytes involved in such responses. Recent studies have revealed nonrandom localization of innate and adaptive immune cells within these organs, suggesting that microanatomical positioning optimizes responses involving sparse cooperating cells. Here, we report that the peripheral localization of LN cDC2 dendritic cells specialized for MHC-II antigen presentation is matched by a similarly biased paracortical distribution of CD4+ T cells directed by the chemoattractant receptor Ebi2. In the absence of Ebi2, CD4+ T cells lose their location bias and are delayed in antigen recognition, proliferative expansion, differentiation, direct effector activity, and provision of help for CD8+ T cell-mediated memory responses, limiting host defense and vaccine responses. These findings demonstrate evolutionary selection for distinct niches within the LN that promote cellular responses, emphasizing the critical link between fine-grained tissue organization and host defense.


Subject(s)
Adaptive Immunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Animals , Antigen Presentation/immunology , Antigens/immunology , Cell Differentiation/immunology , Histocompatibility Antigens Class II/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics
5.
J Immunol ; 208(8): 1845-1850, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35379746

ABSTRACT

Inhibitory receptors have a critical role in the regulation of immunity. Siglecs are a family of primarily inhibitory receptors expressed by immune cells that recognize specific sialic acid modifications on cell surface glycans. Many tumors have increased sialic acid incorporation. Overexpression of the sialyltransferase ST8Sia6 on tumors led to altered immune responses and increased tumor growth. In this study, we examined the role of ST8Sia6 on immune cells in regulating antitumor immunity. ST8Sia6 knockout mice had an enhanced immune response to tumors. The loss of ST8Sia6 promoted an enhanced intratumoral activation of macrophages and dendritic cells, including upregulation of CD40. Intratumoral regulatory T cells exhibited a more inflammatory phenotype in ST8Sia6 knockout mice. Using adoptive transfer studies, the change in regulatory T cell phenotype was not cell intrinsic and depended on the loss of ST8Sia6 expression in APCs. Thus, ST8Sia6 generates ligands for Siglecs that dampen antitumor immunity.


Subject(s)
Neoplasms , Sialyltransferases , Animals , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , N-Acetylneuraminic Acid/immunology , Neoplasms/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sialyltransferases/genetics , Sialyltransferases/immunology
6.
Genes Cells ; 26(3): 180-189, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33527666

ABSTRACT

TRA98 is a rat monoclonal antibody (mAb) which recognizes a specific antigen in the nuclei of germ cells. mAb TRA98 has been used to understand the mechanism of germ cell development and differentiation in many studies. In mice, the antigen recognized by mAb TRA98 or GCNA1 has been reported to be a GCNA gene product, but despite the demonstration of the immunoreactivity of this mAb in human testis and sperm in 1997, the antigen in humans remains unknown, as of date. To identify the human antigen recognized by mAb TRA98, a human comprehensive wet protein array was developed containing 19,446 proteins derived from human cDNAs. Using this array, it was found that the antigen of mAb TRA98 is not a GCNA gene product, but nuclear factor-κB activating protein (NKAP). In mice, mAb TRA98 recognized both the GCNA gene product and NKAP. Furthermore, conditional knockout of Nkap in mice revealed a phenotype of Sertoli cell-only syndrome. Although NKAP is a ubiquitously expressed protein, NKAP recognized by mAb TRA98 in mouse testis was SUMOylated. These results suggest that NKAP undergoes modifications, such as SUMOylation in the testis, and plays an important role in spermatogenesis.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens/metabolism , Germ Cells/metabolism , Protein Array Analysis , Animals , Humans , Male , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , Testis/metabolism
7.
J Immunol ; 204(12): 3071-3076, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32350083

ABSTRACT

The immune system contains a series of checks and balances that maintain tolerance and prevent autoimmunity. Sialic acid-binding Ig-type lectins (Siglecs) are cell surface receptors found on immune cells and inhibit inflammation by recruiting protein tyrosine phosphatases to ITIMs. Islet-resident macrophages express Siglec-E, and Siglec-E expression decreases on islet-resident macrophages as insulitis progresses in the NOD mouse. The sialyltransferase ST8Sia6 generates α-2,8-disialic acids that are ligands for Siglec-E in vivo. We hypothesized that engaging Siglec-E through ST8Sia6-generated ligands may inhibit the development of immune-mediated diabetes. Constitutive overexpression of ST8Sia6 in pancreatic ß cells mitigated hyperglycemia in the multiple low-dose streptozotocin model of diabetes, demonstrating that engagement of this immune receptor facilitates tolerance in the setting of inflammation and autoimmune disease.


Subject(s)
Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Sialyltransferases/metabolism , Streptozocin/pharmacology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Autoimmunity/immunology , Diabetes Mellitus/immunology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Female , Humans , Hyperglycemia/immunology , Hyperglycemia/metabolism , Immune Tolerance/immunology , Inflammation/immunology , Inflammation/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Ligands , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Sialic Acid Binding Ig-like Lectin 2/immunology , Sialic Acid Binding Ig-like Lectin 2/metabolism , Sialyltransferases/immunology
8.
J Immunol ; 202(4): 1033-1038, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30626694

ABSTRACT

Intricate life-versus-death decisions are programmed during T cell development, and the regulatory mechanisms that coordinate their activation and repression are still under investigation. In this study, HDAC3-deficient double-positive (DP) thymocytes exhibit a severe decrease in numbers. The thymic cortex is rich in ATP, which is released by macrophages that clear apoptotic DP thymocytes that fail to undergo positive selection. We demonstrate that HDAC3 is required to repress expression of the purinergic receptor P2X7 to prevent DP cell death. HDAC3-deficient DP thymocytes upregulate the P2X7 receptor, increasing sensitivity to ATP-induced cell death. P2rx7/HDAC3-double knockout mice show a partial rescue in DP cell number. HDAC3 directly binds to the P2rx7 enhancer, which is hyperacetylated in the absence of HDAC3. In addition, RORγt binds to the P2rx7 enhancer and promotes P2X7 receptor expression in the absence of HDAC3. Therefore, HDAC3 is a critical regulator of DP thymocyte survival and is required to suppress P2X7 receptor expression.


Subject(s)
Cell Death , Histone Deacetylases/metabolism , Receptors, Purinergic P2X7/metabolism , Thymocytes/cytology , Thymocytes/enzymology , Animals , Histone Deacetylases/deficiency , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Purinergic P2X7/genetics , Thymocytes/metabolism
9.
J Immunol ; 202(8): 2287-2295, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30804042

ABSTRACT

NKAP is a multifunctional nuclear protein that associates with the histone deacetylase HDAC3. Although both NKAP and HDAC3 are critical for hematopoietic stem cell (HSC) maintenance and survival, it was not known whether these two proteins work together. To assess the importance of their association in vivo, serial truncation and alanine scanning was performed on NKAP to identify the minimal binding site for HDAC3. Mutation of either Y352 or F347 to alanine abrogated the association of NKAP with HDAC3, but did not alter NKAP localization or expression. Using a linked conditional deletion/re-expression system in vivo, we demonstrated that re-expression of the Y352A NKAP mutant failed to restore HSC maintenance and survival in mice when endogenous NKAP expression was eliminated using Mx1-cre and poly-IC, whereas re-expression of wild type NKAP maintained the HSC pool. However, Y352A NKAP did restore proliferation in murine embryonic fibroblasts when endogenous NKAP expression was eliminated using ER-cre and tamoxifen. Therefore, Y352 in NKAP is critical for association with HDAC3 and for HSC maintenance and survival but is not important for proliferation of murine embryonic fibroblasts, demonstrating that NKAP functions in different complexes in different cell types.


Subject(s)
Hematopoietic Stem Cells/immunology , Histone Deacetylases/immunology , Repressor Proteins/immunology , Amino Acid Substitution , Animals , Cell Survival/genetics , Cell Survival/immunology , Embryo, Mammalian/cytology , Embryo, Mammalian/immunology , Fibroblasts/cytology , Fibroblasts/immunology , HEK293 Cells , Hematopoietic Stem Cells/cytology , Histone Deacetylases/genetics , Humans , Mice , Mice, Transgenic , Mutation, Missense , Repressor Proteins/genetics
10.
J Immunol ; 203(2): 408-417, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31175160

ABSTRACT

Recent thymic emigrants that fail postpositive selection maturation are targeted by complement proteins. T cells likely acquire complement resistance during maturation in the thymus, a complement-privileged organ. To test this, thymocytes and fresh serum were separately obtained and incubated together in vitro to assess complement deposition. Complement binding decreased with development and maturation. Complement binding decreased from the double-positive thymocyte to the single-positive stage, and within single-positive thymocytes, complement binding gradually decreased with increasing intrathymic maturation. Binding of the central complement protein C3 to wild-type immature thymocytes required the lectin but not the classical pathway. Specifically, MBL2 but not MBL1 was required, demonstrating a unique function for MBL2. Previous studies demonstrated that the loss of NKAP, a transcriptional regulator of T cell maturation, caused peripheral T cell lymphopenia and enhanced complement susceptibility. To determine whether complement causes NKAP-deficient T cell disappearance, both the lectin and classical pathways were genetically ablated. This blocked C3 deposition on NKAP-deficient T cells but failed to restore normal cellularity, indicating that complement contributes to clearance but is not the primary cause of peripheral T cell lymphopenia. Rather, the accumulation of lipid peroxides in NKAP-deficient T cells was observed. Lipid peroxidation is a salient feature of ferroptosis, an iron-dependent nonapoptotic cell death. Thus, wild-type thymocytes naturally acquire the ability to protect themselves from complement targeting by MBL2 with maturation. However, NKAP-deficient immature peripheral T cells remain scarce in complement-deficient mice likely due to ferroptosis.


Subject(s)
Cell Differentiation/immunology , Complement C3/immunology , Mannose-Binding Lectin/immunology , Repressor Proteins/immunology , T-Lymphocytes/immunology , Animals , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Thymocytes/immunology , Thymus Gland/immunology , Transcription, Genetic/immunology
11.
J Autoimmun ; 89: 139-148, 2018 05.
Article in English | MEDLINE | ID: mdl-29366602

ABSTRACT

Regulatory T cells are critical for the generation and maintenance of peripheral tolerance. Conditional deletion of the transcriptional repressor NKAP in Tregs using Foxp3-YFP-cre NKAP conditional knockout mice causes aggressive autoimmunity characterized by thymic atrophy, lymphadenopathy, peripheral T cell activation, generation of autoantibodies, immune infiltration into several organs, and crusty skin at 3 weeks of age, similar to that of "scurfy" Foxp3-mutant mice. While Treg development in the thymus proceeds normally in the absence of NKAP, there is a severe loss of thymically-derived Tregs in the periphery. NKAP-deficient Tregs have a recent thymic emigrant phenotype, and are attacked by complement in a cell-intrinsic manner in the periphery. Previously, we demonstrated that NKAP is required for conventional T cell maturation as it prevents complement-mediated attack in the periphery. We now show that Tregs undergo a similar maturation process as conventional T cells, requiring NKAP to acquire complement resistance after thymic egress.


Subject(s)
Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/pathology , Animals , Autoantibodies/metabolism , Autoimmunity/genetics , Cell Differentiation , Cells, Cultured , Clonal Deletion , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lymphocyte Activation , Male , Mice , Mice, Knockout , Repressor Proteins/genetics
12.
Immunity ; 30(5): 696-707, 2009 May.
Article in English | MEDLINE | ID: mdl-19409814

ABSTRACT

T cell development depends on the coordinated interplay between receptor signaling and transcriptional regulation. Through a genetic complementation screen a transcriptional repressor, NKAP, was identified. NKAP associated with the histone deacetylase HDAC3 and was shown to be part of a DNA-binding complex, as demonstrated by chromatin immunoprecipitation. NKAP also associated with the Notch corepressor complex. The expression of NKAP during T cell development inversely correlated with the expression of Notch target genes, implying that NKAP may modulate Notch-mediated transcription. To examine the function of NKAP in T cell development, we ablated NKAP by Lck(cre). Loss of NKAP blocked development of alphabeta but not gammadelta T cells, and Nkap(fl/o)Lck(cre) DP T cells expressed 8- to 20-fold higher amounts of Hes1, Deltex1, and CD25 mRNA. Thus, NKAP functions as a transcriptional repressor, acting on Notch target genes, and is required for alphabeta T cell development.


Subject(s)
Gene Expression Regulation , Nuclear Proteins/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Co-Repressor Proteins , Histone Deacetylases/metabolism , Humans , Jurkat Cells , Male , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Repressor Proteins/genetics
13.
J Immunol ; 196(12): 4987-98, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183586

ABSTRACT

Invariant NKT (iNKT) cells are a unique lineage with characteristics of both adaptive and innate lymphocytes, and they recognize glycolipids presented by an MHC class I-like CD1d molecule. During thymic development, iNKT cells also differentiate into NKT1, NKT2, and NKT17 functional subsets that preferentially produce cytokines IFN-γ, IL-4, and IL-17, respectively, upon activation. Newly selected iNKT cells undergo a burst of proliferation, which is defective in mice with a specific deletion of NKAP in the iNKT cell lineage, leading to severe reductions in thymic and peripheral iNKT cell numbers. The decreased cell number is not due to defective homeostasis or increased apoptosis, and it is not rescued by Bcl-xL overexpression. NKAP is also required for differentiation into NKT17 cells, but NKT1 and NKT2 cell development and function are unaffected. This failure in NKT17 development is rescued by transgenic expression of promyelocytic leukemia zinc finger; however, the promyelocytic leukemia zinc finger transgene does not restore iNKT cell numbers or the block in positive selection into the iNKT cell lineage in CD4-cre NKAP conditional knockout mice. Therefore, NKAP regulates multiple steps in iNKT cell development and differentiation.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Natural Killer T-Cells/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Repressor Proteins/metabolism , Animals , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-4/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , bcl-X Protein/genetics
14.
J Immunol ; 197(2): 541-54, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27279370

ABSTRACT

To generate functional peripheral T cells, proper gene regulation during T cell development is critical. In this study, we found that histone deacetylase (HDAC) 3 is required for T cell development. T cell development in CD2-icre HDAC3 conditional knockout (cKO) mice (HDAC3-cKO) was blocked at positive selection, resulting in few CD4 and CD8 T cells, and it could not be rescued by a TCR transgene. These single-positive thymocytes failed to upregulate Bcl-2, leading to increased apoptosis. HDAC3-cKO mice failed to downregulate retinoic acid-related orphan receptor (ROR) γt during positive selection, similar to the block in positive selection in RORγt transgenic mice. In the absence of HDAC3, the RORC promoter was hyperacetylated. In the periphery, the few CD4 T cells present were skewed toward RORγt(+) IL-17-producing Th17 cells, leading to inflammatory bowel disease. Positive selection of CD8 single-positive thymocytes was restored in RORγt-KO Bcl-xL transgenic HDAC3-cKO mice, demonstrating that HDAC3 is required at positive selection to downregulate RORγt.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Histone Deacetylases/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Thymocytes/cytology , Animals , Chromatin Immunoprecipitation , Down-Regulation , Flow Cytometry , Histone Deacetylases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Thymocytes/immunology
15.
J Immunol ; 197(10): 3751-3753, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798172

ABSTRACT

In 2001, The American Association of Immunologists Committee on the Status of Women conducted a survey examining the percentage of women faculty members within immunology departments or women in immunology graduate programs across 27 institutions in the United States, comparing it to the percentage of women receiving a Ph.D. Here, we examine the representation of women across these same 27 immunology departments and programs to examine changes in gender equity over the last 15 years.


Subject(s)
Academies and Institutes/statistics & numerical data , Allergy and Immunology , Education, Graduate , Faculty/statistics & numerical data , Universities , Women , Allergy and Immunology/education , Female , Humans , United States , Workforce
16.
Immunity ; 28(3): 425-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328742

ABSTRACT

Costimulatory signals are critical to T cell activation, but how their effects are mediated remains incompletely characterized. Here, we demonstrate that locally produced C5a and C3a anaphylatoxins interacting with their G protein-coupled receptors (GPCRs), C5aR and C3aR, on APCs and T cells both upstream and downstream of CD28 and CD40L signaling are integrally involved in T cell proliferation and differentiation. Disabling these interactions reduced MHC class II and costimulatory-molecule expression and dramatically diminished T cell responses. Importantly, impaired T cell activation by Cd80-/-Cd86-/- and Cd40-/- APCs was reconstituted by added C5a or C3a. C5aR and C3aR mediated their effects via PI-3 kinase-gamma-dependent AKT phosphorylation, providing a link between GPCR signaling, CD28 costimulation, and T cell survival. These local paracrine and autocrine interactions thus operate constitutively in naive T cells to maintain viability, and their amplification by cognate APC partners thus is critical to T cell costimulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Complement C3a/immunology , Complement C5a/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cell Differentiation/immunology , Complement C3a/metabolism , Complement C5a/metabolism , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Polymerase Chain Reaction , RNA, Messenger/analysis , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction/immunology
17.
J Immunol ; 195(4): 1351-7, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26254267

ABSTRACT

Single-positive thymocytes that successfully complete positive and negative selection must still undergo one final step, generally termed T cell maturation, before they gain functional competency and enter the long-lived T cell pool. Maturation initiates after positive selection in single-positive thymocytes and continues in the periphery in recent thymic emigrants, before these newly produced T cells gain functional competency and are ready to participate in the immune response as peripheral naive T cells. Recent work using genetically altered mice demonstrates that T cell maturation is not a single process, but a series of steps that occur independently and sequentially after positive selection. This review focuses on the changes that occur during T cell maturation, as well as the molecules and pathways that are critical at each step.


Subject(s)
Receptors, Death Domain/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Thymocytes/cytology , Thymocytes/physiology , Animals , Cell Differentiation/immunology , Cell Division , Cell Survival/genetics , Cell Survival/immunology , Clonal Selection, Antigen-Mediated , Humans , Signal Transduction
18.
J Immunol ; 195(4): 1578-90, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163592

ABSTRACT

Recent thymic emigrants are newly generated T cells that need to undergo postthymic maturation to gain functional competency and enter the long-lived naive T cell pool. The mechanism of T cell maturation remains incompletely understood. Previously, we demonstrated that the transcriptional repressor NKAP is required for T cell maturation. Because NKAP associates with histone deacetylase 3 (HDAC3), we examined whether HDAC3 is also required for T cell maturation. Although thymic populations are similar in CD4-cre HDAC3 conditional knockout mice compared with wild-type mice, the peripheral numbers of CD4(+) and CD8(+) T cells are dramatically decreased. In the periphery, the majority of HDAC3-deficient naive T cells are recent thymic emigrants, indicating a block in T cell maturation. CD55 upregulation during T cell maturation is substantially decreased in HDAC3-deficient T cells. Consistent with a block in functional maturation, HDAC3-deficient peripheral T cells have a defect in TNF licensing after TCR/CD28 stimulation. CD4-cre HDAC3 conditional knockout mice do not have a defect in intrathymic migration, thymic egress, T cell survival, or homeostasis. In the periphery, similar to immature NKAP-deficient peripheral T cells, HDAC3-deficient peripheral T cells were bound by IgM and complement proteins, leading to the elimination of these cells. In addition, HDAC3-deficient T cells display decreases in the sialic acid modifications on the cell surface that recruit natural IgM to initiate the classical complement pathway. Therefore, HDAC3 is required for T cell maturation.


Subject(s)
Cell Differentiation , Histone Deacetylases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Histone Deacetylases/genetics , Homeostasis , Interleukin-7/metabolism , Lymphocyte Count , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Tumor Necrosis Factors/metabolism
19.
J Immunol ; 193(12): 6005-15, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25367120

ABSTRACT

Recent thymic emigrants (RTEs) must undergo phenotypic and functional maturation to become long-lived mature naive T cells. In CD4-cre NKAP conditional knockout mice, NKAP-deficient RTEs fail to complete T cell maturation. In this study, we demonstrate that NKAP-deficient immature RTEs do not undergo apoptosis, but are eliminated by complement. C3, C4, and C1q are bound to NKAP-deficient peripheral T cells, demonstrating activation of the classical arm of the complement pathway. As thymocytes mature and exit to the periphery, they increase sialic acid incorporation into cell surface glycans. This is essential to peripheral lymphocyte survival, as stripping sialic acid with neuraminidase leads to the binding of natural IgM and complement fixation. NKAP-deficient T cells have a defect in sialylation on cell surface glycans, leading to IgM recruitment. We demonstrate that the defect in sialylation is due to aberrant α2,8-linked sialylation, and the expression of three genes (ST8sia1, ST8sia4, and ST8sia6) that mediate α2,8 sialylation are downregulated in NKAP-defcient RTEs. The maturation of peripheral NKAP-deficient T cells is partially rescued in a C3-deficient environment. Thus, sialylation during T cell maturation is critical to protect immature RTEs from complement in the periphery.


Subject(s)
Cell Movement/immunology , Complement System Proteins/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Apoptosis/genetics , Apoptosis/immunology , CD55 Antigens/genetics , CD55 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/immunology , Complement Activation/immunology , Complement C3/deficiency , Complement C3/genetics , Complement C3/immunology , Complement System Proteins/metabolism , Gene Expression , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Immunophenotyping , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Knockout , N-Acetylneuraminic Acid/metabolism , Phenotype , Protein Binding/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism
20.
bioRxiv ; 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38463988

ABSTRACT

During the humoral immune response, B cells undergo rapid metabolic reprogramming with a high demand for nutrients, which are vital to sustain the formation of the germinal centers (GCs). Rag-GTPases sense amino acid availability to modulate the mechanistic target of rapamycin complex 1 (mTORC1) pathway and suppress transcription factor EB (TFEB) and transcription factor enhancer 3 (TFE3), members of the microphthalmia (MiT/TFE) family of HLH-leucine zipper transcription factors. However, how Rag-GTPases coordinate amino acid sensing, mTORC1 activation, and TFEB/TFE3 activity in humoral immunity remains undefined. Here, we show that B cell-intrinsic Rag-GTPases are critical for the development and activation of B cells. RagA/RagB deficient B cells fail to form GCs, produce antibodies, and generate plasmablasts in both T-dependent (TD) and T-independent (TI) humoral immune responses. Deletion of RagA/RagB in GC B cells leads to abnormal dark zone (DZ) to light zone (LZ) ratio and reduced affinity maturation. Mechanistically, the Rag-GTPase complex constrains TFEB/TFE3 activity to prevent mitophagy dysregulation and maintain mitochondrial fitness in B cells, which are independent of canonical mTORC1 activation. TFEB/TFE3 deletion restores B cell development, GC formation in Peyer's patches and TI humoral immunity, but not TD humoral immunity in the absence of Rag-GTPases. Collectively, our data establish Rag-GTPase-TFEB/TFE3 pathway as an mTORC1 independent mechanism to coordinating nutrient sensing and mitochondrial metabolism in B cells.

SELECTION OF CITATIONS
SEARCH DETAIL