Your browser doesn't support javascript.
loading
The neonatal microenvironment programs innate γδ T cells through the transcription factor STAT5.
Kadekar, Darshana; Agerholm, Rasmus; Rizk, John; Neubauer, Heidi A; Suske, Tobias; Maurer, Barbara; Viñals, Monica Torrellas; Comelli, Elena M; Taibi, Amel; Moriggl, Richard; Bekiaris, Vasileios.
Afiliação
  • Kadekar D; Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
  • Agerholm R; Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
  • Rizk J; Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
  • Neubauer HA; Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.
  • Suske T; Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.
  • Maurer B; Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.
  • Viñals MT; Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
  • Comelli EM; Department of Nutritional Sciences and.
  • Taibi A; Department of Nutritional Sciences and Joannah and Brian Lawson Centre for Child Nutrition, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
  • Moriggl R; Department of Nutritional Sciences and.
  • Bekiaris V; Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.
J Clin Invest ; 130(5): 2496-2508, 2020 05 01.
Article em En | MEDLINE | ID: mdl-32281944
ABSTRACT
IL-17-producing RORγt+ γδ T cells (γδT17 cells) are innate lymphocytes that participate in type 3 immune responses during infection and inflammation. Herein, we show that γδT17 cells rapidly proliferate within neonatal lymph nodes and gut, where, upon entry, they upregulate T-bet and coexpress IL-17, IL-22, and IFN-γ in a STAT3- and retinoic acid-dependent manner. Neonatal expansion was halted in mice conditionally deficient in STAT5, and its loss resulted in γδT17 cell depletion from all adult organs. Hyperactive STAT5 mutant mice showed that the STAT5A homolog had a dominant role over STAT5B in promoting γδT17 cell expansion and downregulating gut-associated T-bet. In contrast, STAT5B preferentially expanded IFN-γ-producing γδ populations, implying a previously unknown differential role of STAT5 gene products in lymphocyte lineage regulation. Importantly, mice lacking γδT17 cells as a result of STAT5 deficiency displayed a profound resistance to experimental autoimmune encephalomyelitis. Our data identify that the neonatal microenvironment in combination with STAT5 is critical for post-thymic γδT17 development and tissue-specific imprinting, which is essential for infection and autoimmunity.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T / Receptores de Antígenos de Linfócitos T gama-delta / Fator de Transcrição STAT5 / Microambiente Celular / Imunidade Inata / Intestinos Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T / Receptores de Antígenos de Linfócitos T gama-delta / Fator de Transcrição STAT5 / Microambiente Celular / Imunidade Inata / Intestinos Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article