Your browser doesn't support javascript.
loading
Myeloid cell-specific deletion of epidermal growth factor receptor aggravates acute cardiac injury.
Okyere, Ama D; Nayak, Tapas K; Patwa, Viren; Teplitsky, David; McEachern, Erin; Carter, Rhonda L; Xu, Heli; Gao, Erhe; Zhou, Yan; Tilley, Douglas G.
Afiliação
  • Okyere AD; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Nayak TK; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Patwa V; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Teplitsky D; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • McEachern E; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Carter RL; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Xu H; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Gao E; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
  • Zhou Y; Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, U.S.A.
  • Tilley DG; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, U.S.A.
Clin Sci (Lond) ; 137(19): 1513-1531, 2023 10 11.
Article em En | MEDLINE | ID: mdl-37728308
ABSTRACT
Myeloid cells, including macrophages, play important roles as first responders to cardiac injury and stress. Epidermal growth factor receptor (EGFR) has been identified as a mediator of macrophage responsiveness to select diseases, though its impact on cardiac function or remodeling following acute ischemic injury is unknown. We aimed to define the role of myeloid cell-specific EGFR in the regulation of cardiac function and remodeling following acute myocardial infarction (MI)-induced injury. Floxed EGFR mice were bred with homozygous LysM-Cre (LMC) transgenic mice to yield myeloid-specific EGFR knockout (mKO) mice. Via echocardiography, immunohistochemistry, RNA sequencing and flow cytometry, the impact of myeloid cell-specific EGFR deletion on cardiac structure and function was assessed at baseline and following injury. Compared with LMC controls, myeloid cell-specific EGFR deletion led to an increase in cardiomyocyte hypertrophy at baseline. Bulk RNASeq analysis of isolated cardiac Cd11b+ myeloid cells revealed substantial changes in mKO cell transcripts at baseline, particularly in relation to predicted decreases in neovascularization. In response to myocardial infarction, mKO mice experienced a hastened decline in cardiac function with isolated cardiac Cd11b+ myeloid cells expressing decreased levels of the pro-reparative mediators Vegfa and Il10, which coincided with enhanced cardiac hypertrophy and decreased capillary density. Overall, loss of EGFR qualitatively alters cardiac resident macrophages that promotes a low level of basal stress and a more rapid decrease in cardiac function along with worsened repair following acute ischemic injury.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores ErbB / Infarto do Miocárdio Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores ErbB / Infarto do Miocárdio Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2023 Tipo de documento: Article