Your browser doesn't support javascript.
loading
Crosstalk between cytotoxic CD8+ T cells and stressed cardiomyocytes triggers development of interstitial cardiac fibrosis in hypertensive mouse hearts.
Brassington, Kurt; Kanellakis, Peter; Cao, Anh; Toh, Ban-Hock; Peter, Karlheinz; Bobik, Alex; Kyaw, Tin.
Afiliação
  • Brassington K; Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
  • Kanellakis P; Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
  • Cao A; Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
  • Toh BH; Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.
  • Peter K; Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.
  • Bobik A; Inflammation and Cardiovascular Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
  • Kyaw T; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia.
Front Immunol ; 13: 1040233, 2022.
Article em En | MEDLINE | ID: mdl-36483558
ABSTRACT

Aims:

Cardiac fibrosis is central to heart failure (HF), especially HF with preserved ejection fraction (HFpEF), often caused by hypertension. Despite fibrosis causing diastolic dysfunction and impaired electrical conduction, responsible for arrhythmia-induced sudden cardiac death, the mechanisms are poorly defined and effective therapies are lacking. Here we show that crosstalk between cardiac cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is essential for development of non-ischemic hypertensive cardiac fibrosis. Methods and

results:

CD8 T cell depletion in hypertensive mice, strongly attenuated CF, reduced cardiac apoptosis and improved ventricular relaxation. Interaction between cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is highly dependent on the CD8+ T cells expressing the innate stress-sensing receptor NKG2D and stressed cardiomyocytes expressing the NKG2D activating ligand RAE-1. The interaction between NKG2D and RAE-1 results in CD8+ T cell activation, release of perforin, cardiomyocyte apoptosis, increased numbers of TGF-ß1 expressing macrophages and fibrosis. Deleting NKG2D or perforin from CD8+ T cells greatly attenuates these effects. Activation of the cytoplasmic DNA-STING-TBK1-IRF3 signaling pathway in overly stressed cardiomyocytes is responsible for elevating RAE-1 and MCP-1, a macrophage attracting chemokine. Inhibiting STING activation greatly attenuates cardiomyocyte RAE-1 expression, the cardiomyocyte apoptosis, TGF-ß1 and fibrosis.

Conclusion:

Our data highlight a novel pathway by which CD8 T cells contribute to an early triggering mechanism in CF development; preventing CD8+ T cell activation by inhibiting the cardiomyocyte RAE-1-CD8+ T cell-NKG2D axis holds promise for novel therapeutic strategies to limit hypertensive cardiac fibrosis.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Insuficiência Cardíaca / Hipertensão Tipo de estudo: Etiology_studies Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Insuficiência Cardíaca / Hipertensão Tipo de estudo: Etiology_studies Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article