Your browser doesn't support javascript.
loading
Epigenetic Networks Regulate the Transcriptional Program in Memory and Terminally Differentiated CD8+ T Cells.
Rodriguez, Ramon M; Suarez-Alvarez, Beatriz; Lavín, José L; Mosén-Ansorena, David; Baragaño Raneros, Aroa; Márquez-Kisinousky, Leonardo; Aransay, Ana M; Lopez-Larrea, Carlos.
Afiliación
  • Rodriguez RM; Department of Immunology, Central University Hospital of Asturias, 33011 Oviedo, Spain.
  • Suarez-Alvarez B; Department of Immunology, Central University Hospital of Asturias, 33011 Oviedo, Spain.
  • Lavín JL; Genome Analysis Platform, CIC bioGUNE and CIBERehd, Technological Park of Bizkaia, 48160 Derio, Spain.
  • Mosén-Ansorena D; Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA 02215; and.
  • Baragaño Raneros A; Department of Immunology, Central University Hospital of Asturias, 33011 Oviedo, Spain.
  • Márquez-Kisinousky L; Department of Immunology, Central University Hospital of Asturias, 33011 Oviedo, Spain.
  • Aransay AM; Genome Analysis Platform, CIC bioGUNE and CIBERehd, Technological Park of Bizkaia, 48160 Derio, Spain.
  • Lopez-Larrea C; Department of Immunology, Central University Hospital of Asturias, 33011 Oviedo, Spain; inmuno@hca.es.
J Immunol ; 198(2): 937-949, 2017 01 15.
Article en En | MEDLINE | ID: mdl-27974453
ABSTRACT
Epigenetic mechanisms play a critical role during differentiation of T cells by contributing to the formation of stable and heritable transcriptional patterns. To better understand the mechanisms of memory maintenance in CD8+ T cells, we performed genome-wide analysis of DNA methylation, histone marking (acetylated lysine 9 in histone H3 and trimethylated lysine 9 in histone), and gene-expression profiles in naive, effector memory (EM), and terminally differentiated EM (TEMRA) cells. Our results indicate that DNA demethylation and histone acetylation are coordinated to generate the transcriptional program associated with memory cells. Conversely, EM and TEMRA cells share a very similar epigenetic landscape. Nonetheless, the TEMRA transcriptional program predicts an innate immunity phenotype associated with genes never reported in these cells, including several mediators of NK cell activation (VAV3 and LYN) and a large array of NK receptors (e.g., KIR2DL3, KIR2DL4, KIR2DL1, KIR3DL1, KIR2DS5). In addition, we identified up to 161 genes that encode transcriptional regulators, some of unknown function in CD8+ T cells, and that were differentially expressed in the course of differentiation. Overall, these results provide new insights into the regulatory networks involved in memory CD8+ T cell maintenance and T cell terminal differentiation.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Diferenciación Celular / Regulación de la Expresión Génica / Linfocitos T CD8-positivos / Epigénesis Genética / Memoria Inmunológica Límite: Humans Idioma: En Revista: J Immunol Año: 2017 Tipo del documento: Article País de afiliación: España

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Diferenciación Celular / Regulación de la Expresión Génica / Linfocitos T CD8-positivos / Epigénesis Genética / Memoria Inmunológica Límite: Humans Idioma: En Revista: J Immunol Año: 2017 Tipo del documento: Article País de afiliación: España