Neutrophil-derived S100A8/A9 amplify granulopoiesis after myocardial infarction

G Sreejit, A Abdel-Latif, B Athmanathan… - Circulation, 2020 - Am Heart Assoc
G Sreejit, A Abdel-Latif, B Athmanathan, R Annabathula, A Dhyani, SK Noothi
Circulation, 2020Am Heart Assoc
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 …
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-1β secretion. The released interleukin-1β 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-1β) in neutrophils suppress granulopoiesis and may improve cardiac function in patients with acute coronary syndrome.
Am Heart Assoc