Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 329
Filtrar
1.
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
2.
Annu Rev Immunol ; 38: 421-453, 2020 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31990619

RESUMO

Foxp3-expressing CD4+ regulatory T (Treg) cells play key roles in the prevention of autoimmunity and the maintenance of immune homeostasis and represent a major barrier to the induction of robust antitumor immune responses. Thus, a clear understanding of the mechanisms coordinating Treg cell differentiation is crucial for understanding numerous facets of health and disease and for developing approaches to modulate Treg cells for clinical benefit. Here, we discuss current knowledge of the signals that coordinate Treg cell development, the antigen-presenting cell types that direct Treg cell selection, and the nature of endogenous Treg cell ligands, focusing on evidence from studies in mice. We also highlight recent advances in this area and identify key unanswered questions.


Assuntos
Diferenciação Celular/imunologia , Linfopoese/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Biomarcadores , Diferenciação Celular/genética , Deleção Clonal , Seleção Clonal Mediada por Antígeno , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Linfopoese/genética , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Timo/citologia , Timo/imunologia , Timo/metabolismo
3.
Nat Immunol ; 19(12): 1366-1378, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420627

RESUMO

Thymocyte development requires a complex orchestration of multiple transcription factors. Ablating either TCF-1 or HEB in CD4+CD8+ thymocytes elicits similar developmental outcomes including increased proliferation, decreased survival, and fewer late Tcra rearrangements. Here, we provide a mechanistic explanation for these similarities by showing that TCF-1 and HEB share ~7,000 DNA-binding sites genome wide and promote chromatin accessibility. The binding of both TCF-1 and HEB was required at these shared sites for epigenetic and transcriptional gene regulation. Binding of TCF-1 and HEB to their conserved motifs in the enhancer regions of genes associated with T cell differentiation promoted their expression. Binding to sites lacking conserved motifs in the promoter regions of cell-cycle-associated genes limited proliferation. TCF-1 displaced nucleosomes, allowing for chromatin accessibility. Importantly, TCF-1 inhibited Notch signaling and consequently protected HEB from Notch-mediated proteasomal degradation. Thus, TCF-1 shifts nucleosomes and safeguards HEB, thereby enabling their cooperation in establishing the epigenetic and transcription profiles of CD4+CD8+ thymocytes.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Regulação da Expressão Gênica/imunologia , Fator 1-alfa Nuclear de Hepatócito/imunologia , Linfopoese/imunologia , Timócitos/imunologia , Animais , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
Nat Immunol ; 19(12): 1427-1440, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30374131

RESUMO

Multipotent progenitor cells confirm their T cell-lineage identity in the CD4-CD8- double-negative (DN) pro-T cell DN2 stages, when expression of the essential transcription factor Bcl11b begins. In vivo and in vitro stage-specific deletions globally identified Bcl11b-controlled target genes in pro-T cells. Proteomics analysis revealed that Bcl11b associated with multiple cofactors and that its direct action was needed to recruit those cofactors to selective target sites. Regions near functionally regulated target genes showed enrichment for those sites of Bcl11b-dependent recruitment of cofactors, and deletion of individual cofactors relieved the repression of many genes normally repressed by Bcl11b. Runx1 collaborated with Bcl11b most frequently for both activation and repression. In parallel, Bcl11b indirectly regulated a subset of target genes by a gene network circuit via the transcription inhibitor Id2 (encoded by Id2) and transcription factor PLZF (encoded by Zbtb16); Id2 and Zbtb16 were directly repressed by Bcl11b, and Id2 and PLZF controlled distinct alternative programs. Thus, our study defines the molecular basis of direct and indirect Bcl11b actions that promote T cell identity and block alternative potentials.


Assuntos
Linfopoese/imunologia , Células Precursoras de Linfócitos T/imunologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/biossíntese , Proteínas Repressoras/imunologia , Proteínas Supressoras de Tumor/imunologia , Animais , Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Precursoras de Linfócitos T/citologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia
5.
Nat Immunol ; 18(9): 973-984, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28671690

RESUMO

The balance of myeloid populations and lymphoid populations must be well controlled. Here we found that osteopontin (OPN) skewed this balance during pathogenic conditions such as infection and autoimmunity. Notably, two isoforms of OPN exerted distinct effects in shifting this balance through cell-type-specific regulation of apoptosis. Intracellular OPN (iOPN) diminished the population size of myeloid progenitor cells and myeloid cells, and secreted OPN (sOPN) increase the population size of lymphoid cells. The total effect of OPN on skewing the leukocyte population balance was observed as host sensitivity to early systemic infection with Candida albicans and T cell-mediated colitis. Our study suggests previously unknown detrimental roles for two OPN isoforms in causing the imbalance of leukocyte populations.


Assuntos
Doenças Autoimunes/imunologia , Candidíase/imunologia , Colite/imunologia , Infecções/imunologia , Linfócitos/imunologia , Células Mieloides/imunologia , Osteopontina/imunologia , Animais , Apoptose , Candida albicans , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Linfopoese/imunologia , Camundongos , Camundongos Knockout , Mielopoese/imunologia , Osteopontina/genética , Isoformas de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T
6.
Immunity ; 51(5): 930-948.e6, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31604687

RESUMO

Generation of the first T lymphocytes in the human embryo involves the emergence, migration, and thymus seeding of lymphoid progenitors together with concomitant thymus organogenesis, which is the initial step to establish the entire adaptive immune system. However, the cellular and molecular programs regulating this process remain unclear. We constructed a single-cell transcriptional landscape of human early T lymphopoiesis by using cells from multiple hemogenic and hematopoietic sites spanning embryonic and fetal stages. Among heterogenous early thymic progenitors, one subtype shared common features with a subset of lymphoid progenitors in fetal liver that are known as thymus-seeding progenitors. Unbiased bioinformatics analysis identified a distinct type of pre-thymic lymphoid progenitors in the aorta-gonad-mesonephros (AGM) region. In parallel, we investigated thymic epithelial cell development and potential cell-cell interactions during thymus organogenesis. Together, our data provide insights into human early T lymphopoiesis that prospectively direct T lymphocyte regeneration, which might lead to development of clinical applications.


Assuntos
Diferenciação Celular/genética , Linfopoese/genética , Organogênese/genética , Células Precursoras de Linfócitos T/citologia , Células Precursoras de Linfócitos T/metabolismo , Timo/embriologia , Biomarcadores , Diferenciação Celular/imunologia , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Linfopoese/imunologia , Detecção de Sinal Psicológico , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timo/imunologia , Timo/metabolismo , Transcriptoma
7.
Immunity ; 48(6): 1119-1134.e7, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29924977

RESUMO

Transcription factors normally regulate gene expression through their action at sites where they bind to DNA. However, the balance of activating and repressive functions that a transcription factor can mediate is not completely understood. Here, we showed that the transcription factor PU.1 regulated gene expression in early T cell development both by recruiting partner transcription factors to its own binding sites and by depleting them from the binding sites that they preferred when PU.1 was absent. The removal of partner factors Satb1 and Runx1 occurred primarily from sites where PU.1 itself did not bind. Genes linked to sites of partner factor "theft" were enriched for genes that PU.1 represses despite lack of binding, both in a model cell line system and in normal T cell development. Thus, system-level competitive recruitment dynamics permit PU.1 to affect gene expression both through its own target sites and through action at a distance.


Assuntos
Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Linfopoese/fisiologia , Proteínas Proto-Oncogênicas/imunologia , Linfócitos T/imunologia , Transativadores/imunologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Linfopoese/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
8.
Nat Immunol ; 15(6): 492-502, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24840980

RESUMO

Immunological reactions are propelled by ever-changing signals that alter the translational ability of the RNA in the cells involved. Such alterations are considered to be consequential modifications in the transcriptomic decoding of the genetic blueprint. The identification of RNA-binding protein (RBP) assemblies engaged in the coordinative regulation of state-specific RNAs indicates alternative and exclusive means for determining the activation, plasticity and tolerance of cells of the immune system. Here we review current knowledge about RBP-regulated post-transcriptional events involved in the reactivity of cells of the immune system and the importance of their alteration during chronic inflammatory pathology and autoimmunity.


Assuntos
Imunidade Celular/genética , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Ribonucleico/genética , Animais , Autoimunidade/genética , Autoimunidade/imunologia , Humanos , Tolerância Imunológica/genética , Imunidade Celular/imunologia , Inflamação/genética , Inflamação/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Linfopoese/genética , Linfopoese/imunologia , Camundongos , Proteínas de Ligação a RNA/genética , Sequências Reguladoras de Ácido Ribonucleico/imunologia , Transdução de Sinais/imunologia
9.
Nat Immunol ; 15(3): 283-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24509509

RESUMO

The transcription factor Ikaros is an essential regulator of lymphopoiesis. Here we studied its B cell-specific function by conditional inactivation of the gene encoding Ikaros (Ikzf1) in pro-B cells. B cell development was arrested at an aberrant 'pro-B cell' stage characterized by increased cell adhesion and loss of signaling via the pre-B cell signaling complex (pre-BCR). Ikaros activated genes encoding signal transducers of the pre-BCR and repressed genes involved in the downregulation of pre-BCR signaling and upregulation of the integrin signaling pathway. Unexpectedly, derepression of expression of the transcription factor Aiolos did not compensate for the loss of Ikaros in pro-B cells. Ikaros induced or suppressed active chromatin at regulatory elements of activated or repressed target genes. Notably, binding of Ikaros and expression of its target genes were dynamically regulated at distinct stages of early B lymphopoiesis.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Fator de Transcrição Ikaros/imunologia , Linfopoese/imunologia , Células Precursoras de Linfócitos B/citologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Imunoprecipitação da Cromatina , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Fator de Transcrição Ikaros/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/metabolismo
10.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35072209

RESUMO

Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα-/- environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Linfopoese/genética , Receptores de Interleucina-7/genética , Tirosina Quinase 3 Semelhante a fms/genética , Imunidade Adaptativa/genética , Animais , Linfócitos B/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Hematopoéticas/citologia , Imunidade Inata/genética , Linfócitos/citologia , Linfócitos/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Linfopoese/imunologia , Camundongos , Especificidade de Órgãos/genética , Linfócitos T Reguladores/imunologia
11.
Nat Immunol ; 14(8): 867-75, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23812095

RESUMO

The transcription factors EBF1 and Pax5 have been linked to activation of the B cell lineage program and irreversible loss of alternative lineage potential (commitment), respectively. Here we conditionally deleted Ebf1 in committed pro-B cells after transfer into alymphoid mice. We found that those cells converted into innate lymphoid cells (ILCs) and T cells with variable-diversity-joining (VDJ) rearrangements of loci encoding both B cell and T cell antigen receptors. As intermediates in lineage conversion, Ebf1-deficient CD19(+) cells expressing Pax5 and transcriptional regulators of the ILC and T cell fates were detectable. In particular, genes encoding the transcription factors Id2 and TCF-1 were bound and repressed by EBF1. Thus, both EBF1 and Pax5 are required for B lineage commitment by repressing distinct and common determinants of alternative cell fates.


Assuntos
Linfócitos B/imunologia , Transativadores/imunologia , Transferência Adotiva , Animais , Linfócitos B/citologia , Diferenciação Celular/imunologia , Linhagem da Célula , DNA/química , DNA/genética , Regulação da Expressão Gênica , Linfopoese/imunologia , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Transativadores/genética , Recombinação V(D)J/genética , Recombinação V(D)J/imunologia
12.
Nat Immunol ; 14(12): 1277-84, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24185616

RESUMO

Notch signaling induces gene expression of the T cell lineage and discourages alternative fate outcomes. Hematopoietic deficiency in the Notch target Hes1 results in severe T cell lineage defects; however, the underlying mechanism is unknown. We found here that Hes1 constrained myeloid gene-expression programs in T cell progenitor cells, as deletion of the myeloid regulator C/EBP-α restored the development of T cells from Hes1-deficient progenitor cells. Repression of Cebpa by Hes1 required its DNA-binding and Groucho-recruitment domains. Hes1-deficient multipotent progenitor cells showed a developmental bias toward myeloid cells and dendritic cells after Notch signaling, whereas Hes1-deficient lymphoid progenitor cells required additional cytokine signaling for diversion into the myeloid lineage. Our findings establish the importance of constraining developmental programs of the myeloid lineage early in T cell development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Proteína alfa Estimuladora de Ligação a CCAAT/imunologia , Proteínas de Homeodomínio/imunologia , Receptor Notch1/imunologia , Linfócitos T/imunologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Linhagem Celular , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Linfopoese/genética , Linfopoese/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Células Mieloides/imunologia , Células Mieloides/metabolismo , Ligação Proteica/imunologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Linfócitos T/metabolismo , Fatores de Transcrição HES-1
13.
Immunity ; 45(3): 527-539, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27566938

RESUMO

B cell development is often depicted as a linear process initiating in the fetus and continuing postnatally. Using a PU.1 hypomorphic mouse model, we found that B-1 and B-2 lymphopoiesis occurred in distinct fetal and adult waves differentially dependent on the Sfpi1 14 kB upstream regulatory element. The initial wave of fetal B-1 development was absent in PU.1 hypomorphic mice, while subsequent fetal and adult waves emerged. In contrast, B-2 lymphopoiesis occurred in distinct fetal and adult waves. Whole-transcriptome profiling of fetal and adult B cell progenitors supported the existence of three waves of B-1 and two waves of B-2 development and revealed that the network of transcription factors governing B lineage specification and commitment was highly divergent between B-1 and B-2 progenitors. These findings support the view that the B-1 and B-2 lineages are distinct and provide a genetic basis for layering of immune system development.


Assuntos
Subpopulações de Linfócitos B/imunologia , Redes Reguladoras de Genes/imunologia , Linfopoese/imunologia , Animais , Linhagem da Célula/imunologia , Feto/imunologia , Perfilação da Expressão Gênica/métodos , Camundongos , Células Precursoras de Linfócitos B/imunologia , Fatores de Transcrição/imunologia
14.
Nat Immunol ; 12(5): 434-40, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21423172

RESUMO

B lymphopoiesis begins in the fetal liver, switching after birth to the bone marrow, where it persists for life. The unique developmental outcomes of each phase are well documented, yet their molecular requirements are not. Here we describe two allelic X-linked mutations in mice that caused cell-intrinsic arrest of adult B lymphopoiesis. Mutant fetal liver progenitors generated B cells in situ but not in irradiated adult bone marrow, which emphasizes a necessity for the affected pathway only in the context of adult bone marrow. The causative mutations were ascribed to Atp11c, which encodes a P4-type ATPase with no previously described function. Our data establish an essential, cell-autonomous and context-sensitive function for ATP11C, a putative aminophospholipid flippase, in B cell development.


Assuntos
Adenosina Trifosfatases/imunologia , Linfócitos B/imunologia , Medula Óssea/imunologia , Linfopoese/imunologia , Adenosina Trifosfatases/genética , Animais , Linfócitos B/enzimologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Imunofenotipagem , Interleucina-7/imunologia , Linfopoese/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transativadores/imunologia
15.
Blood ; 138(4): 304-317, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-33786586

RESUMO

Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited because of a lack of methods to isolate these cells. We found that murine Lin-CD31-Sca-1-CD51+ cells can be divided into 4 subpopulations by using flow cytometry based on their expression of the platelet-derived growth factor receptors ⍺ and ß (PDGFR⍺ and PDGFRß). The use of different skeletal lineage reporters confirmed the skeletal origin of the 4 populations. Multiplex immunohistochemistry studies revealed that all 4 populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit-fibroblast capacity, and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the 4 populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that 1 of these 4 different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B-lymphocyte precursors were also observed in these distinct skeletal sites. This cell population also supported pre-B lymphopoiesis in culture. Our method of isolating 4 distinct maturing skeletal populations will help elucidate the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular/imunologia , Linfopoese/imunologia , Músculo Esquelético/imunologia , Animais , Diferenciação Celular/genética , Linfopoese/genética , Camundongos , Camundongos Transgênicos
16.
Eur J Immunol ; 51(5): 1080-1088, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33521937

RESUMO

TCRαß+ CD8α+ CD8ß- intestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely ß2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+ . However, selective depletion of ß2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.


Assuntos
Seleção Clonal Mediada por Antígeno , Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/metabolismo , Linfopoese , Células Precursoras de Linfócitos T/imunologia , Células Precursoras de Linfócitos T/metabolismo , Timo/citologia , Animais , Biomarcadores , Diferenciação Celular , Genes MHC Classe I , Imunofenotipagem , Linfócitos Intraepiteliais/citologia , Linfopoese/genética , Linfopoese/imunologia , Camundongos , Células Precursoras de Linfócitos T/citologia , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo
17.
Nat Immunol ; 11(2): 171-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19946273

RESUMO

STAT5 and interleukin 7 (IL-7) signaling are thought to control B lymphopoiesis by regulating the expression of key transcription factors and by activating variable (V(H)) gene segments at the immunoglobulin heavy-chain (Igh) locus. Using conditional mutagenesis to delete the gene encoding the transcription factor STAT5, we demonstrate that the development of pro-B cells was restored by transgenic expression of the prosurvival protein Bcl-2, which compensated for loss of the antiapoptotic protein Mcl-1. Expression of the genes encoding the B cell-specification factor EBF1 and the B cell-commitment protein Pax5 as well as V(H) gene recombination were normal in STAT5- or IL-7 receptor alpha-chain (IL-7Ralpha)-deficient pro-B cells rescued by Bcl-2. STAT5-expressing pro-B cells contained little or no active chromatin at most V(H) genes. In contrast, rearrangements of the immunoglobulin-kappa light-chain locus (Igk) were more abundant in STAT5- or IL-7Ralpha-deficient pro-B cells. Hence, STAT5 and IL-7 signaling control cell survival and the developmental ordering of immunoglobulin gene rearrangements by suppressing premature Igk recombination in pro-B cells.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/genética , Células Progenitoras Linfoides/citologia , Linfopoese/genética , Fator de Transcrição STAT5/genética , Transdução de Sinais/imunologia , Animais , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Rearranjo Gênico/genética , Rearranjo Gênico/imunologia , Genes de Imunoglobulinas/genética , Genes de Imunoglobulinas/imunologia , Interleucina-7/genética , Interleucina-7/imunologia , Células Progenitoras Linfoides/imunologia , Linfopoese/imunologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/imunologia
18.
Stem Cells ; 39(4): 389-402, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33400834

RESUMO

Hematopoietic stem cells (HSCs) are the progenitor cells that give rise to the diverse repertoire of all immune cells. As they differentiate, HSCs yield a series of cell states that undergo gradual commitment to become mature blood cells. Studies of hematopoiesis in murine models have provided critical insights about the lineage relationships among stem cells, progenitors, and mature cells, and these have guided investigations of the molecular basis for these distinct developmental stages. Primary immune deficiencies are caused by inborn errors of immunity that result in immune dysfunction and subsequent susceptibility to severe and recurrent infection(s). Over the last decade there has been a dramatic increase in the number and depth of the molecular, cellular, and clinical characterization of such genetically defined causes of immune dysfunction. Patients harboring inborn errors of immunity thus represent a unique resource to improve our understanding of the multilayered and complex mechanisms underlying lymphocyte development in humans. These breakthrough discoveries not only enable significant advances in the diagnosis of such rare and complex conditions but also provide substantial improvement in the development of personalized treatments. Here, we will discuss the clinical, cellular, and molecular phenotypes, and treatments of selected inborn errors of immunity that impede, either intrinsically or extrinsically, the development of B- or T-cells at different stages.


Assuntos
Regulação da Expressão Gênica/imunologia , Células-Tronco Hematopoéticas/imunologia , Linfopoese/imunologia , Medicina de Precisão/métodos , Doenças da Imunodeficiência Primária/genética , Animais , Diferenciação Celular , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Terapia Genética/métodos , Hematopoese/genética , Hematopoese/imunologia , Células-Tronco Hematopoéticas/citologia , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/imunologia , Janus Quinase 3/genética , Janus Quinase 3/imunologia , Linfócitos/imunologia , Linfócitos/patologia , Linfopoese/genética , Camundongos , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/imunologia , Doenças da Imunodeficiência Primária/patologia , Doenças da Imunodeficiência Primária/terapia
19.
Immunity ; 38(5): 943-57, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23623382

RESUMO

Macrophage and dendritic cell (DC) progenitors (MDPs) and common DC progenitors (CDPs) are bone marrow (BM) progenitors with DC differentiation potential. However, both MDPs and CDPs give rise to large numbers of conventional DCs (cDCs) and few plasmacytoid DCs (pDCs), implying that more dedicated pDC progenitors remain to be identified. Here we have described DC progenitors with a prominent pDC differentiation potential. Although both MDPs and CDPs express the macrophage colony stimulating factor (M-CSF) receptor (M-CSFR), the progenitors were confined to a M-CSFR(-) fraction, identified as Lin(-)c-Kit(int/lo)Flt3(+)M-CSFR(-), and expressed high amounts of E2-2 (also known as Tcf4) an essential transcription factor for pDC development. Importantly, they appeared to be directly derived from either CDPs or lymphoid-primed multipotent progenitors (LMPPs). Collectively, our findings provide insight into DC differentiation pathways and may lead to progenitor-based therapeutic applications for infection and autoimmune disease.


Assuntos
Células Dendríticas/metabolismo , Células Progenitoras Linfoides/metabolismo , Linfopoese/imunologia , Macrófagos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Células da Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Células Progenitoras Linfoides/imunologia , Linfopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Fator Estimulador de Colônias de Macrófagos/biossíntese , Receptor de Fator Estimulador de Colônias de Macrófagos/deficiência , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator de Transcrição 4 , Tirosina Quinase 3 Semelhante a fms/metabolismo
20.
J Immunol ; 204(9): 2447-2454, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32198141

RESUMO

The caudal hematopoietic tissue in zebrafish, the equivalent to the fetal liver in mammals, is an intermediate hematopoietic niche for the maintenance and differentiation of hematopoietic stem and progenitor cells before homing to the thymus and kidney marrow. As one of the ultimate hematopoietic organs, the thymus sustains T lymphopoiesis, which is essential for adaptive immune system. However, the mechanism of prethymic T lymphoid progenitors migrating to the thymus remains elusive. In this study, we identify an Rho GTPase Rac2 as a modulator of T lymphoid progenitor homing to the thymus in zebrafish. rac2-Deficient embryos show the inability of T lymphoid progenitors homing to the thymus because of defective cell-autonomous motility. Mechanistically, we demonstrate that Rac2 regulates homing of T lymphoid progenitor through Pak1-mediated AKT pathway. Taken together, our work reveals an important function of Rac2 in directing T lymphoid progenitor migration to the thymus during zebrafish embryogenesis.


Assuntos
Movimento Celular/imunologia , Desenvolvimento Embrionário/imunologia , Células Progenitoras Linfoides/metabolismo , Timo/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados/imunologia , Animais Geneticamente Modificados/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Células Progenitoras Linfoides/imunologia , Linfopoese/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Timo/imunologia , Peixe-Zebra/imunologia , Proteínas de Peixe-Zebra/imunologia , Quinases Ativadas por p21/imunologia , Quinases Ativadas por p21/metabolismo , Proteínas rac de Ligação ao GTP/imunologia , Proteínas rho de Ligação ao GTP/imunologia , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa