Your browser doesn't support javascript.
loading
Deficiency of histone lysine methyltransferase SETDB2 in hematopoietic cells promotes vascular inflammation and accelerates atherosclerosis.
Zhang, Xinbo; Sun, Jonathan; Canfrán-Duque, Alberto; Aryal, Binod; Tellides, George; Chang, Ying Ju; Suárez, Yajaira; Osborne, Timothy F; Fernández-Hernando, Carlos.
Afiliação
  • Zhang X; Vascular Biology and Therapeutics Program.
  • Sun J; Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and.
  • Canfrán-Duque A; Vascular Biology and Therapeutics Program.
  • Aryal B; Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and.
  • Tellides G; Vascular Biology and Therapeutics Program.
  • Chang YJ; Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and.
  • Suárez Y; Vascular Biology and Therapeutics Program.
  • Osborne TF; Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and.
  • Fernández-Hernando C; Vascular Biology and Therapeutics Program.
JCI Insight ; 6(12)2021 06 22.
Article em En | MEDLINE | ID: mdl-34003795
ABSTRACT
Epigenetic modifications of the genome, including DNA methylation, histone methylation/acetylation, and noncoding RNAs, have been reported to play a fundamental role in regulating immune response during the progression of atherosclerosis. SETDB2 is a member of the KMT1 family of lysine methyltransferases, and members of this family typically methylate histone H3 Lys9 (H3K9), an epigenetic mark associated with gene silencing. Previous studies have shown that SETDB2 is involved in innate and adaptive immunity, the proinflammatory response, and hepatic lipid metabolism. Here, we report that expression of SETDB2 is markedly upregulated in human and murine atherosclerotic lesions. Upregulation of SETDB2 was observed in proinflammatory M1 but not antiinflammatory M2 macrophages. Notably, we found that genetic deletion of SETDB2 in hematopoietic cells promoted vascular inflammation and enhanced the progression of atherosclerosis in BM transfer studies in Ldlr-knockout mice. Single-cell RNA-Seq analysis in isolated CD45+ cells from atherosclerotic plaques from mice transplanted with SETDB2-deficient BM revealed a significant increase in monocyte population and enhanced expression of genes involved in inflammation and myeloid cell recruitment. Additionally, we found that loss of SETDB2 in hematopoietic cells was associated with macrophage accumulation in atherosclerotic lesions and attenuated efferocytosis. Overall, these studies identify SETDB2 as an important inflammatory cell regulator that controls macrophage activation in atherosclerotic plaques.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Histona-Lisina N-Metiltransferase / Aterosclerose / Inflamação / Macrófagos Limite: Animals / Humans / Male Idioma: En Revista: JCI Insight Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Histona-Lisina N-Metiltransferase / Aterosclerose / Inflamação / Macrófagos Limite: Animals / Humans / Male Idioma: En Revista: JCI Insight Ano de publicação: 2021 Tipo de documento: Article