Your browser doesn't support javascript.
loading
Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction.
Sreejit, Gopalkrishna; Abdel-Latif, Ahmed; Athmanathan, Baskaran; Annabathula, Rahul; Dhyani, Ashish; Noothi, Sunil K; Quaife-Ryan, Gregory A; Al-Sharea, Annas; Pernes, Gerard; Dragoljevic, Dragana; Lal, Hind; Schroder, Kate; Hanaoka, Beatriz Y; Raman, Chander; Grant, Maria B; Hudson, James E; Smyth, Susan S; Porrello, Enzo R; Murphy, Andrew J; Nagareddy, Prabhakara R.
  • Sreejit G; Department of Surgery (G.S., B.A., P.R.N.), Ohio State University Wexner Medical Center, Columbus.
  • Abdel-Latif A; Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham.
  • Athmanathan B; Department of Medicine, University of Kentucky, Lexington (A.A.-L., R.A., S.S.S.).
  • Annabathula R; Department of Surgery (G.S., B.A., P.R.N.), Ohio State University Wexner Medical Center, Columbus.
  • Dhyani A; Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham.
  • Noothi SK; Department of Medicine, University of Kentucky, Lexington (A.A.-L., R.A., S.S.S.).
  • Quaife-Ryan GA; Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham.
  • Al-Sharea A; Departments of Pathology (G.S., B.A., A.D., S.K.N., P.R.N.), University of Alabama at Birmingham.
  • Pernes G; Ophthalmology and Visual Sciences (S.K.N., M.B.G.), University of Alabama at Birmingham.
  • Dragoljevic D; School of Biomedical Sciences (G.A.Q.-R.), University of Queensland, St. Lucia, Australia.
  • Lal H; QIMR Berghofer Medical Research Institute, Brisbane, Australia (G.A.Q.-R., J.E.H.).
  • Schroder K; Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.).
  • Hanaoka BY; Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.).
  • Raman C; Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia (A.A.-S., G.P., D.D., A.J.M.).
  • Grant MB; Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham.
  • Hudson JE; Institute for Molecular Bioscience (IMB) (K.S.), University of Queensland, St. Lucia, Australia.
  • Smyth SS; IMB Centre for Inflammation and Disease Research (K.S.), University of Queensland, St. Lucia, Australia.
  • Porrello ER; Department of Medicine (B.Y.H.), Ohio State University Wexner Medical Center, Columbus.
  • Murphy AJ; Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham.
  • Nagareddy PR; Medicine (H.L., B.Y.H., C.R.), University of Alabama at Birmingham.
Circulation ; 141(13): 1080-1094, 2020 03 31.
Article en En | MEDLINE | ID: mdl-31941367
ABSTRACT

BACKGROUND:

Myocardial infarction (MI) triggers myelopoiesis, resulting in heightened production of neutrophils. However, the mechanisms that sustain their production and recruitment to the injured heart are unclear.

METHODS:

Using a mouse model of the permanent ligation of the left anterior descending artery and flow cytometry, we first characterized the temporal and spatial effects of MI on different myeloid cell types. We next performed global transcriptome analysis of different cardiac cell types within the infarct to identify the drivers of the acute inflammatory response and the underlying signaling pathways. Using a combination of genetic and pharmacological strategies, we identified the sequelae of events that led to MI-induced myelopoiesis. Cardiac function was assessed by echocardiography. The association of early indexes of neutrophilia with major adverse cardiovascular events was studied in a cohort of patients with acute MI.

RESULTS:

Induction of MI results in rapid recruitment of neutrophils to the infarct, where they release specific alarmins, S100A8 and S100A9. These alarmins bind to the Toll-like receptor 4 and prime the nod-like receptor family pyrin domain-containing 3 inflammasome in naïve neutrophils and promote interleukin-1ß secretion. The released interleukin-1ß interacts with its receptor (interleukin 1 receptor type 1) on hematopoietic stem and progenitor cells in the bone marrow and stimulates granulopoiesis in a cell-autonomous manner. Genetic or pharmacological strategies aimed at disruption of S100A8/A9 and their downstream signaling cascade suppress MI-induced granulopoiesis and improve cardiac function. Furthermore, in patients with acute coronary syndrome, higher neutrophil count on admission and after revascularization correlates positively with major adverse cardiovascular disease outcomes.

CONCLUSIONS:

Our study provides novel evidence for the primary role of neutrophil-derived alarmins (S100A8/A9) in dictating the nature of the ensuing inflammatory response after myocardial injury. Therapeutic strategies aimed at disruption of S100A8/A9 signaling or their downstream mediators (eg, nod-like receptor family pyrin domain-containing 3 inflammasome, interleukin-1ß) in neutrophils suppress granulopoiesis and may improve cardiac function in patients with acute coronary syndrome.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Calgranulina A / Granulocitos / Infarto del Miocardio / Neutrófilos Límite: Animals / Female / Humans / Male Idioma: En Año: 2020 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Calgranulina A / Granulocitos / Infarto del Miocardio / Neutrófilos Límite: Animals / Female / Humans / Male Idioma: En Año: 2020 Tipo del documento: Article