Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Semin Immunol ; 66: 101709, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36621291

RESUMO

Innate lymphoid cells (ILCs) are a group of innate lymphocytes that do not express RAG-dependent rearranged antigen-specific cell surface receptors. ILCs are classified into five groups according to their developmental trajectory and cytokine production profile. They encompass NK cells, which are cytotoxic, helper-like ILCs 1-3, which functionally mirror CD4+ T helper (Th) type 1, Th2 and Th17 cells respectively, and lymphoid tissue inducer (LTi) cells. NK cell development depends on Eomes (eomesodermin), whereas the ILC1 program is regulated principally by the transcription factor T-bet (T-box transcription factor Tbx21), that of ILC2 is regulated by GATA3 (GATA-binding protein 3) and that of ILC3 is regulated by RORγt (RAR-related orphan receptor γ). NK cells were discovered close to fifty years ago, but ILC1s were first described only about fifteen years ago. Within the ILC family, NK and ILC1s share many similarities, as witnessed by their cell surface phenotype which largely overlap. NK cells and ILC1s have been reported to respond to tissue inflammation and intracellular pathogens. Several studies have reported an antitumorigenic role for NK cells in both humans and mice, but data for ILC1s are both scarce and contradictory. In this review, we will first describe the different NK cell and ILC1 subsets, their effector functions and development. We will then discuss their role in cancer and the effects of the tumor microenvironment on their metabolism.


Assuntos
Imunidade Inata , Células Matadoras Naturais , Linfócitos , Neoplasias , Animais , Humanos , Camundongos , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Neoplasias/imunologia , Linfócitos T Auxiliares-Indutores , Microambiente Tumoral
2.
Cell Rep Med ; 3(11): 100812, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36384102

RESUMO

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1s) are populations of non-T, non-B lymphocytes in peripheral tissues. Although NK and ILC1 subsets have been described, their identification and characteristics remain unclear. We performed single-cell RNA sequencing and CITE-seq to explore NK and ILC1 heterogeneity between tissues. We observed that although NK1 and NK2 subsets are conserved in spleen and liver, ILC1s are heterogeneous across tissues. We identified sets of genes expressed by related subsets or characterizing unique ILC1 populations in each organ. The syndecan-4 appeared as a marker discriminating murine ILC1 from NK cells across organs. Finally, we revealed that the expressions of EOMES, GZMA, IRF8, JAK1, NKG7, PLEK, PRF1, and ZEB2 define NK cells and that IL7R, LTB, and RGS1 differentiate ILC1s from NK cells in mice and humans. Our data constitute an important resource to improve our understanding of NK-ILC1 origin, phenotype, and biology.


Assuntos
Imunidade Inata , Células Matadoras Naturais , Animais , Humanos , Camundongos , Imunidade Inata/genética , Células Matadoras Naturais/metabolismo
3.
Elife ; 112022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35188458

RESUMO

Interactions of developing T cells with Aire+ medullary thymic epithelial cells expressing high levels of MHCII molecules (mTEChi) are critical for the induction of central tolerance in the thymus. In turn, thymocytes regulate the cellularity of Aire+ mTEChi. However, it remains unknown whether thymocytes control the precursors of Aire+ mTEChi that are contained in mTEClo cells or other mTEClo subsets that have recently been delineated by single-cell transcriptomic analyses. Here, using three distinct transgenic mouse models, in which antigen presentation between mTECs and CD4+ thymocytes is perturbed, we show by high-throughput RNA-seq that self-reactive CD4+ thymocytes induce key transcriptional regulators in mTEClo and control the composition of mTEClo subsets, including Aire+ mTEChi precursors, post-Aire and tuft-like mTECs. Furthermore, these interactions upregulate the expression of tissue-restricted self-antigens, cytokines, chemokines, and adhesion molecules important for T-cell development. This gene activation program induced in mTEClo is combined with a global increase of the active H3K4me3 histone mark. Finally, we demonstrate that these self-reactive interactions between CD4+ thymocytes and mTECs critically prevent multiorgan autoimmunity. Our genome-wide study thus reveals that self-reactive CD4+ thymocytes control multiple unsuspected facets from immature stages of mTECs, which determines their heterogeneity.


Assuntos
Autoantígenos/fisiologia , Células Epiteliais/fisiologia , Timócitos/fisiologia , Timo , Animais , Linfócitos T CD4-Positivos , Proteínas de Ligação a DNA , Epitélio/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Histonas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso , Transdução de Sinais
4.
Nature ; 594(7864): 501-502, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34079112
5.
Nat Immunol ; 22(2): 179-192, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33462452

RESUMO

Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias do Colo/metabolismo , Metabolismo Energético , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Subpopulações de Linfócitos T/metabolismo , Timo/metabolismo , Microambiente Tumoral , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Linhagem da Célula , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Feminino , Glucose/metabolismo , Glicólise , Humanos , Imunoterapia Adotiva , Interferon gama/metabolismo , Interleucina-17/metabolismo , Metabolismo dos Lipídeos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/transplante , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Obesidade/imunologia , Obesidade/metabolismo , Técnicas de Cultura de Órgãos , Fenótipo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante , Timo/imunologia , Carga Tumoral
6.
Eur J Immunol ; 51(1): 17-26, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33188652

RESUMO

γδ T cells can display a plethora of immune functions, but recent studies have highlighted their importance, in multiple disease models, as sources of the pro-inflammatory cytokines, IL-17A (IL-17), and IFN-γ. These are produced by distinct murine effector γδ T cell subsets that diverge during thymic γδ T cell development. Among the multiple roles these subsets play in peripheral tissues, a striking dichotomy has emerged at tumor sites: whereas IFN-γ+ γδ T cells inhibit tumor cell growth, IL-17+ γδ T cells promote tumor progression and metastasis formation. In this review, we discuss the main lines of evidence, mostly from preclinical studies in mouse models, for this functional dichotomy in cancer immunity. We further highlight very recent advances in our understanding how metabolic sources and pathways can impact on the balance between IFN-γ+ and IL-17+ γδ T cells in the tumor microenvironment, which opens a new exciting avenue to explore toward the application of γδ T cells in cancer immunotherapy.


Assuntos
Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/metabolismo , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Animais , Proliferação de Células , Citotoxicidade Imunológica , Humanos , Interferon gama/metabolismo , Interleucina-17/metabolismo , Camundongos , Modelos Imunológicos , Células Supressoras Mieloides/imunologia , Neoplasias Experimentais/patologia , Neovascularização Patológica , Microambiente Tumoral/imunologia
7.
Nat Rev Immunol ; 21(4): 221-232, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33057185

RESUMO

γδ T cells are a unique T cell subpopulation that are rare in secondary lymphoid organs but enriched in many peripheral tissues, such as the skin, intestines and lungs. By rapidly producing large amounts of cytokines, γδ T cells make key contributions to immune responses in these tissues. In addition to their immune surveillance activities, recent reports have unravelled exciting new roles for γδ T cells in steady-state tissue physiology, with functions ranging from the regulation of thermogenesis in adipose tissue to the control of neuronal synaptic plasticity in the central nervous system. Here, we review the roles of γδ T cells in tissue homeostasis and in surveillance of infection, aiming to illustrate their major impact on tissue integrity, tissue repair and immune protection.


Assuntos
Vigilância Imunológica/fisiologia , Infecções/imunologia , Linfócitos Intraepiteliais/fisiologia , Mucosa/fisiologia , Plasticidade Neuronal/fisiologia , Regeneração/fisiologia , Termogênese/fisiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiologia , Regeneração Óssea/fisiologia , Butirofilinas/metabolismo , Sistema Nervoso Central/fisiologia , Feminino , Genitália Feminina/fisiologia , Gengiva/fisiologia , Homeostase , Humanos , Vigilância Imunológica/imunologia , Mucosa Intestinal/fisiologia , Linfócitos Intraepiteliais/imunologia , Pulmão/fisiologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/fisiologia
8.
Nat Commun ; 9(1): 1262, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29593265

RESUMO

Medullary thymic epithelial cells (mTEC) purge the T cell repertoire of autoreactive thymocytes. Although dendritic cells (DC) reinforce this process by transporting innocuous peripheral self-antigens, the mechanisms that control their thymic entry remain unclear. Here we show that mTEC-CD4+ thymocyte crosstalk regulates the thymus homing of SHPS-1+ conventional DCs (cDC), plasmacytoid DCs (pDC) and macrophages. This homing process is controlled by lymphotoxin α (LTα), which negatively regulates CCL2, CCL8 and CCL12 chemokines in mTECs. Consequently, Ltα-deficient mice have increased expression of these chemokines that correlates with augmented classical NF-κB subunits and increased thymic recruitment of cDCs, pDCs and macrophages. This enhanced migration depends mainly on the chemokine receptor CCR2, and increases thymic clonal deletion. Altogether, this study identifies a fine-tuning mechanism of T cell repertoire selection and paves the way for therapeutic interventions to treat autoimmune disorders.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Deleção Clonal , Linfotoxina-alfa/metabolismo , Timo/imunologia , Animais , Antígenos/imunologia , Células da Medula Óssea/imunologia , Quimiocinas/imunologia , Técnicas de Cocultura , Células Dendríticas/imunologia , Feminino , Deleção de Genes , Tolerância Imunológica , Ligantes , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , NF-kappa B/metabolismo , Receptores CCR2/metabolismo , Linfócitos T/imunologia , Timócitos/imunologia
9.
EMBO Mol Med ; 9(6): 835-851, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28455312

RESUMO

Cytoablative treatments lead to severe damages on thymic epithelial cells (TECs), which result in delayed de novo thymopoiesis and a prolonged period of T-cell immunodeficiency. Understanding the mechanisms that govern thymic regeneration is of paramount interest for the recovery of a functional immune system notably after bone marrow transplantation (BMT). Here, we show that RANK ligand (RANKL) is upregulated in CD4+ thymocytes and lymphoid tissue inducer (LTi) cells during the early phase of thymic regeneration. Importantly, whereas RANKL neutralization alters TEC recovery after irradiation, ex vivo RANKL administration during BMT boosts the regeneration of TEC subsets including thymic epithelial progenitor-enriched cells, thymus homing of lymphoid progenitors, and de novo thymopoiesis. RANKL increases specifically in LTi cells, lymphotoxin α, which is critical for thymic regeneration. RANKL treatment, dependent on lymphotoxin α, is beneficial upon BMT in young and aged individuals. This study thus indicates that RANKL may be clinically useful to improve T-cell function recovery after BMT by controlling multiple facets of thymic regeneration.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Células Epiteliais/fisiologia , Ligante RANK/administração & dosagem , Radioterapia/efeitos adversos , Regeneração , Timo/fisiologia , Animais , Linfotoxina-alfa/metabolismo , Camundongos , Resultado do Tratamento
10.
Med Sci (Paris) ; 31(8-9): 742-7, 2015.
Artigo em Francês | MEDLINE | ID: mdl-26340833

RESUMO

The establishment of thymic central tolerance is a critical process to prevent the development of autoimmune diseases. Medullary thymic epithelial cells (mTEC) are essential to this process through the expression of the transcription factor Aire, which controls the transcription of many genes encoding tissue-restricted antigens. Mutations in the Aire gene are responsible for a rare autoimmune disorder called APECED (autoimmune polyendocrinopathy candidiasis ectodermal dystrophy). This review summarizes our current knowledge on the mode of action of Aire at the molecular and epigenetic levels in controlling the expression of tissue-restricted antigens. We also discuss recently described additional roles of this transcription factor in the induction of central T-cell tolerance.


Assuntos
Tolerância Central/genética , Epigênese Genética , Fatores de Transcrição/genética , Humanos , Proteína AIRE
11.
Front Immunol ; 6: 365, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257733

RESUMO

The thymus ensures the generation of a functional and highly diverse T-cell repertoire. The thymic medulla, which is mainly composed of medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), provides a specialized microenvironment dedicated to the establishment of T-cell tolerance. mTECs play a privileged role in this pivotal process by their unique capacity to express a broad range of peripheral self-antigens that are presented to developing T cells. Reciprocally, developing T cells control mTEC differentiation and organization. These bidirectional interactions are commonly referred to as thymic crosstalk. This review focuses on the relative contributions of mTEC and DC subsets to the deletion of autoreactive T cells and the generation of natural regulatory T cells. We also summarize current knowledge regarding how hematopoietic cells conversely control the composition and complex three-dimensional organization of the thymic medulla.

12.
Cell Rep ; 10(1): 39-46, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25543139

RESUMO

In cancer, immune cells can play conflicting roles, either protective, by elimination of tumor cells during immune surveillance, or detrimental, by promoting carcinogenesis during inflammation. We report here that the thymus-specific serine protease (TSSP), which is involved in CD4(+) T cell maturation in the thymus, exerts a tumor suppressor activity. Mice genetically deficient for TSSP are highly prone to spontaneous cancer development. The absence of TSSP also increases the rate of induced colitis-associated colorectal (CAC) tumor formation, through exacerbated colon inflammation. Adoptive transfer of T cells in various combinations (CD4(+) and CD8(+) from wild-type and/or knockout mice) into T cell-deficient mice showed that the TSSP-deficient CD4(+) T cell compartment promotes tumor development, associated with high levels of the cytokine IL-17A. Inhibition of IL-17A during CAC tumor formation prevents the increased carcinogenesis and colic immune disequilibrium observed in TSSP-deficient mice. Therefore, our data demonstrate that antitumoral immune surveillance requires thymic TSSP-driven production of CD4(+) T cells contributing to inflammatory balance.


Assuntos
Colite/genética , Neoplasias Colorretais/genética , Inflamação/genética , Serina Endopeptidases/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Colite/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Knockout , Serina Endopeptidases/biossíntese , Timo/imunologia , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA