Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38677292

RESUMO

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.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Glicólise , Imunidade Inata , Linfócitos , Camundongos Knockout , Animais , Camundongos , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética , Hexoquinase/metabolismo , Hexoquinase/genética , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Interleucina-17/metabolismo , Adaptação Fisiológica/imunologia
2.
J Immunol ; 208(8): 1845-1850, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35379746

RESUMO

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.


Assuntos
Neoplasias , Sialiltransferases , Animais , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/imunologia , Ácido N-Acetilneuramínico/imunologia , Neoplasias/imunologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Sialiltransferases/genética , Sialiltransferases/imunologia
3.
Elife ; 102021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34854376

RESUMO

After antigenic activation, quiescent naive CD4+ T cells alter their metabolism to proliferate. This metabolic shift increases production of nucleotides, amino acids, fatty acids, and sterols. Here, we show that histone deacetylase 3 (HDAC3) is critical for activation of murine peripheral CD4+ T cells. HDAC3-deficient CD4+ T cells failed to proliferate and blast after in vitro TCR/CD28 stimulation. Upon T-cell activation, genes involved in cholesterol biosynthesis are upregulated while genes that promote cholesterol efflux are repressed. HDAC3-deficient CD4+ T cells had reduced levels of cellular cholesterol both before and after activation. HDAC3-deficient cells upregulate cholesterol synthesis appropriately after activation, but fail to repress cholesterol efflux; notably, they overexpress cholesterol efflux transporters ABCA1 and ABCG1. Repression of these genes is the primary function for HDAC3 in peripheral CD4+ T cells, as addition of exogenous cholesterol restored proliferative capacity. Collectively, these findings demonstrate HDAC3 is essential during CD4+ T-cell activation to repress cholesterol efflux.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Colesterol/metabolismo , Histona Desacetilases/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Colesterol/genética , Feminino , Histona Desacetilases/genética , Masculino , Camundongos , Camundongos Mutantes
4.
Elife ; 102021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762046

RESUMO

Iron-sulfur (Fe-S) clusters are cofactors essential for the activity of numerous enzymes including DNA polymerases, helicases, and glycosylases. They are synthesized in the mitochondria as Fe-S intermediates and are exported to the cytoplasm for maturation by the mitochondrial transporter ABCB7. Here, we demonstrate that ABCB7 is required for bone marrow B cell development, proliferation, and class switch recombination, but is dispensable for peripheral B cell homeostasis in mice. Conditional deletion of ABCB7 using Mb1-cre resulted in a severe block in bone marrow B cell development at the pro-B cell stage. The loss of ABCB7 did not alter expression of transcription factors required for B cell specification or commitment. While increased intracellular iron was observed in ABCB7-deficient pro-B cells, this did not lead to increased cellular or mitochondrial reactive oxygen species, ferroptosis, or apoptosis. Interestingly, loss of ABCB7 led to replication-induced DNA damage in pro-B cells, independent of VDJ recombination, and these cells had evidence of slowed DNA replication. Stimulated ABCB7-deficient splenic B cells from CD23-cre mice also had a striking loss of proliferation and a defect in class switching. Thus, ABCB7 is essential for early B cell development, proliferation, and class switch recombination.


Assuntos
Linfócitos B/fisiologia , Switching de Imunoglobulina , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Animais , Proliferação de Células , Dano ao DNA , Feminino , Ferro/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Baço/citologia , Enxofre/metabolismo
5.
Cancer Immunol Res ; 9(8): 952-966, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34074677

RESUMO

Many tumors exhibit increased incorporation of sialic acids into cell-surface glycans, which impact the tumor microenvironment. Sialic acid immunoglobulin-like lectins (Siglec) are receptors that recognize sialic acids and modulate immune responses, including responses to tumors. However, the roles of individual sialyltransferases in tumorigenesis and tumor growth are not well understood. Here, we examined the sialyltransferase ST8Sia6, which generated α2,8-linked disialic acids that bind to murine Siglec-E and human Siglec-7 and -9. Increased ST8Sia6 expression was found on many human tumors and associated with decreased survival in several cancers, including colon cancer. Because of this, we engineered MC38 and B16-F10 tumor lines to express ST8Sia6. ST8Sia6-expressing MC38 and B16-F10 tumors exhibited faster growth and led to decreased survival, which required host Siglec-E. ST8Sia6 expression on tumors also altered macrophage polarization toward M2, including upregulation of the immune modulator arginase, which also required Siglec-E. ST8Sia6 also accelerated tumorigenesis in a genetically engineered, spontaneous murine model of colon cancer, decreasing survival from approximately 6 months to 67 days. Thus, ST8Sia6 expression on tumors inhibits antitumor immune responses to accelerate tumor growth.


Assuntos
Imunidade Inata/imunologia , Imunidade/imunologia , Sialiltransferases/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Transfecção , Microambiente Tumoral
6.
Genes Cells ; 26(3): 180-189, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33527666

RESUMO

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.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos/metabolismo , Células Germinativas/metabolismo , Análise Serial de Proteínas , Animais , Humanos , Masculino , Camundongos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Testículo/metabolismo
7.
Cell Rep ; 34(1): 108601, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33406440

RESUMO

Immune cells can metabolize glucose, amino acids, and fatty acids (FAs) to generate energy. The roles of different FA species and their impacts on humoral immunity remain poorly understood. Here, we report that proliferating B cells require monounsaturated FAs (MUFAs) to maintain mitochondrial metabolism and mTOR activity and to prevent excessive autophagy and endoplasmic reticulum (ER) stress. Furthermore, B cell-extrinsic stearoyl-CoA desaturase (SCD) activity generates MUFA to support early B cell development and germinal center (GC) formation in vivo during immunization and influenza infection. Thus, SCD-mediated MUFA production is critical for humoral immunity.


Assuntos
Linfócitos B/fisiologia , Ácidos Graxos Monoinsaturados/imunologia , Ácidos Graxos Monoinsaturados/metabolismo , Imunidade Humoral , Mitocôndrias/fisiologia , Estearoil-CoA Dessaturase/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia , Estresse do Retículo Endoplasmático , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/imunologia , Serina-Treonina Quinases TOR/imunologia
8.
J Immunol ; 204(12): 3071-3076, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32350083

RESUMO

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.


Assuntos
Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/metabolismo , Sialiltransferases/metabolismo , Estreptozocina/farmacologia , Animais , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Diferenciação de Linfócitos B/metabolismo , Autoimunidade/imunologia , Diabetes Mellitus/imunologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Hiperglicemia/imunologia , Hiperglicemia/metabolismo , Tolerância Imunológica/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Ligantes , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Sialiltransferases/imunologia
9.
Annu Rev Immunol ; 38: 397-419, 2020 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31990620

RESUMO

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.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Linfopoese , Linfócitos T/imunologia , Linfócitos T/metabolismo , Acetilação , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Histonas , Humanos , Linfopoese/genética , Linfopoese/imunologia , Metilação , Processamento de Proteína Pós-Traducional , Linfócitos T/citologia , Linfócitos T/enzimologia , Ubiquitinação
10.
Front Cell Dev Biol ; 7: 214, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31632967

RESUMO

NKAP is a multi-functional nuclear protein that has been shown to be essential for hematopoiesis. Deletion of NKAP in hematopoietic stem cells (HSCs) was previously found to result in rapid lethality and hematopoietic failure. NKAP deficient cells also exhibited diminished proliferation and increased expression of the cyclin dependent kinase inhibitors (CDKIs) p19 Ink4d and p21 Cip1. To determine how dysregulation of CDKI expression contributes to the effects of NKAP deficiency, NKAP was deleted in mice also deficient in p19 Ink4d or p21 Cip1 using poly-IC treatment to induce Mx1-cre. Hematopoietic failure and lethality were not prevented by deficiency in either CDKI when NKAP was deleted. Inducible deletion of NKAP in cultured hematopoietic progenitors ex vivo resulted in a senescent phenotype and altered expression of numerous cell cycle regulators including the CDKI p16 INK4a. Interestingly, while combined deficiency in p16 INK4a and p21 Cip1 did not reverse the effect of NKAP deficiency on hematopoiesis in vivo, it did shift the consequence of NKAP deficiency from senescence to apoptosis in ex vivo cultures. These results suggest that NKAP may limit cellular stress that can trigger cell cycle withdrawal or cell death, a role critical for the maintenance of a viable pool of hematopoietic progenitors.

11.
Immunohorizons ; 3(8): 352-367, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31387873

RESUMO

NKAP and HDAC3 are critical for T cell maturation. NKAP and HDAC3 physically associate, and a point mutation in NKAP, NKAP(Y352A), abrogates this interaction. To evaluate the significance of NKAP and HDAC3 association in T cell maturation, transgenic mice were engineered for cre-mediated endogenous NKAP gene deletion coupled to induction of NKAP(Y352A) or a wild type (WT) control transgene, NKAP(WT), in double positive thymocytes or regulatory T cells (Tregs). T cell maturation was normal in mice with endogenous NKAP deletion coupled to NKAP(WT) induction. However, severe defects occurred in T cell and Treg maturation and in iNKT cell development when NKAP(Y352A) was induced, recapitulating NKAP deficiency. Conventional T cells expressing NKAP(Y352A) failed to enter the long-term T cell pool, did not produce cytokines, and remained complement susceptible, whereas Tregs expressing NKAP(Y352A) were eliminated as recent thymic emigrants leading to lethal autoimmunity. Overall, these results demonstrate the significance of NKAP-HDAC3 association in T cells.


Assuntos
Diferenciação Celular/fisiologia , Histona Desacetilases/metabolismo , Células T Matadoras Naturais/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T Reguladores/metabolismo , Timócitos/metabolismo , Animais , Autoimunidade/genética , Células Cultivadas , Ativação do Complemento , Complemento C3/imunologia , Citocinas/metabolismo , Feminino , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Peroxidação de Lipídeos/genética , Masculino , Camundongos , Camundongos Knockout , Mutação Puntual , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Timo/citologia
12.
J Immunol ; 203(2): 408-417, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31175160

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Complemento C3/imunologia , Lectina de Ligação a Manose/imunologia , Proteínas Repressoras/imunologia , Linfócitos T/imunologia , Animais , Linfopenia/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Timócitos/imunologia , Timo/imunologia , Transcrição Gênica/imunologia
13.
J Immunol ; 202(8): 2287-2295, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30804042

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Histona Desacetilases/imunologia , Proteínas Repressoras/imunologia , Substituição de Aminoácidos , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/imunologia , Fibroblastos/citologia , Fibroblastos/imunologia , Células HEK293 , Células-Tronco Hematopoéticas/citologia , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Proteínas Repressoras/genética
14.
Elife ; 82019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30657451

RESUMO

CD4 and CD8 T cells are vital components of the immune system. We found that histone deacetylase 3 (HDAC3) is critical for the development of CD4 T cells, as HDAC3-deficient DP thymocytes generate only CD8SP thymocytes in mice. In the absence of HDAC3, MHC Class II-restricted OT-II thymocytes are redirected to the CD8 cytotoxic lineage, which occurs with accelerated kinetics. Analysis of histone acetylation and RNA-seq reveals that HDAC3-deficient DP thymocytes are biased towards the CD8 lineage prior to positive selection. Commitment to the CD4 or CD8 lineage is determined by whether persistent TCR signaling or cytokine signaling predominates, respectively. Despite elevated IL-21R/γc/STAT5 signaling in HDAC3-deficient DP thymocytes, blocking IL-21R does not restore CD4 lineage commitment. Instead, HDAC3 binds directly to CD8-lineage promoting genes. Thus, HDAC3 is required to restrain CD8-lineage genes in DP thymocytes for the generation of CD4 T cells.


Assuntos
Antígenos CD4/imunologia , Antígenos CD8/imunologia , Linhagem da Célula/genética , Expressão Gênica/fisiologia , Histona Desacetilases/fisiologia , Timócitos/citologia , Animais , Masculino , Camundongos , Transdução de Sinais , Timócitos/imunologia
15.
J Autoimmun ; 89: 139-148, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29366602

RESUMO

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.


Assuntos
Proteínas Repressoras/metabolismo , Linfócitos T Reguladores/imunologia , Timo/patologia , Animais , Autoanticorpos/metabolismo , Autoimunidade/genética , Diferenciação Celular , Células Cultivadas , Deleção Clonal , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética
16.
Sci Rep ; 7(1): 7018, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765611

RESUMO

iNKT cells are a unique lineage of T cells that recognize glycolipid presented by CD1d. In the thymus, they differentiate into iNKT1, iNKT2 and iNKT17 effector subsets, characterized by preferential expression of Tbet, Gata3 and ROR-γt and production of IFN-γ, IL-4 and IL-17, respectively. We demonstrate that the transcriptional regulator Runx1 is essential for the generation of ROR-γt expressing iNKT17 cells. PLZF-cre Runx1 cKO mice lack iNKT17 cells in the thymus, spleen and liver. Runx1-deficient iNKT cells have altered expression of several genes important for iNKT17 differentiation, including decreased expression of IL-7Rα, BATF and c-Maf and increased expression of Bcl11b and Lef1. However, reduction of Lef1 expression or introduction of an IL-7Rα transgene is not sufficient to correct the defect in iNKT17 differentiation, demonstrating that Runx1 is a key regulator of several genes required for iNKT17 differentiation. Loss of Runx1 leads to a severe decrease in iNKT cell numbers in the thymus, spleen and liver. The decrease in cell number is due to a combined decrease in proliferation at Stage 1 during thymic development and increased apoptosis. Thus, we describe a novel role of Runx1 in iNKT cell development and differentiation, particularly in orchestrating iNKT17 differentiation.


Assuntos
Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células T Matadoras Naturais/fisiologia , Subpopulações de Linfócitos T/fisiologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Fígado/patologia , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/química , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/análise , Baço/patologia , Subpopulações de Linfócitos T/química , Timo/patologia
17.
Sci Rep ; 7(1): 5784, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724935

RESUMO

NKT cells are a distinct subset that have developmental requirements that often differ from conventional T cells. Here, we show that NKT-specific deletion of Hdac3 results in a severe reduction in the number of iNKT cells, particularly of NKT1 cells. In addition, there is decreased cytokine production by Hdac3-deficient NKT2 and NKT17 cells. Hdac3-deficient iNKT cells have increased cell death that is not rescued by transgenic expression of Bcl-2 or Bcl-xL. Hdac3-deficient iNKT cells have less Cyto-ID staining and lower LC3A/B expression, indicative of reduced autophagy. Interestingly, Hdac3-deficient iNKT cells also have lower expression of the nutrient receptors GLUT1, CD71 and CD98, which would increase the need for autophagy when nutrients are limiting. Therefore, Hdac3 is required for iNKT cell development and differentiation.


Assuntos
Diferenciação Celular , Histona Desacetilases/metabolismo , Células T Matadoras Naturais/fisiologia , Animais , Apoptose , Sobrevivência Celular , Citocinas/metabolismo , Deleção de Genes , Histona Desacetilases/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
J Immunol ; 197(10): 3751-3753, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27798172

RESUMO

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.


Assuntos
Academias e Institutos/estatística & dados numéricos , Alergia e Imunologia , Educação de Pós-Graduação , Docentes/estatística & dados numéricos , Universidades , Mulheres , Alergia e Imunologia/educação , Feminino , Humanos , Estados Unidos , Recursos Humanos
19.
J Immunol ; 197(2): 541-54, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27279370

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Histona Desacetilases/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Timócitos/citologia , Animais , Imunoprecipitação da Cromatina , Regulação para Baixo , Citometria de Fluxo , Histona Desacetilases/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Timócitos/imunologia
20.
J Immunol ; 196(12): 4987-98, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27183586

RESUMO

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.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Células T Matadoras Naturais/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Proteínas Repressoras/metabolismo , Animais , Proliferação de Células , Citocinas/biossíntese , Citocinas/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Interleucina-4/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteína bcl-X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...