1. Academic Validation
  2. HMGB1-Promoted Neutrophil Extracellular Traps Contribute to Cardiac Diastolic Dysfunction in Mice

HMGB1-Promoted Neutrophil Extracellular Traps Contribute to Cardiac Diastolic Dysfunction in Mice

  • J Am Heart Assoc. 2022 Feb 15;11(4):e023800. doi: 10.1161/JAHA.121.023800.
Xin-Lin Zhang 1 Ting-Yu Wang 2 3 Zheng Chen 1 Hong-Wei Wang 2 3 Yong Yin 1 Lian Wang 1 Yong Wang 2 3 Biao Xu 1 Wei Xu 1
Affiliations

Affiliations

  • 1 Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China.
  • 2 Central for Translational Medicine Nanjing University School of Medicine Nanjing China.
  • 3 State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine Nanjing University School of Medicine Nanjing China.
Abstract

Background Heart failure with preserved ejection fraction (HFpEF) remains an increasing public health problem with substantial morbidity and mortality but with few effective treatments. A novel inflammatory mechanism has been proposed, but the inflammatory signals promoting the development of HFpEF remain greatly unknown. Methods and Results Serum of patients with HFpEF was collected for measurement of circulating neutrophils and markers of neutrophil extracellular traps (NETs). To induce HFpEF phenotype, male C57BL/6 mice underwent uninephrectomy, received a continuous infusion of d-aldosterone for 4 weeks, and maintained on 1.0% sodium chloride drinking water. Heart tissues were harvested, immune cell types determined by flow cytometry, NETs formation by immunofluorescence, and western blotting. Differentiated neutrophils were cultured to investigate the effect of HMGB1 (high mobility group protein B1) and SGLT2 (sodium-glucose cotransporter-2) inhibitor on NETs formation in vitro. Circulating neutrophils and NETs markers are elevated in patients with HFpEF, as are cardiac neutrophils and NETs formation in HFpEF mice. NETs inhibition with deoxyribonuclease 1 in experimental HFpEF mice reduces heart macrophages infiltration and inflammation and ameliorates cardiac fibrosis and diastolic function. Damage-associated molecular pattern HMGB1 expression is elevated in cardiac tissue of HFpEF mice, and HMGB1 inhibition reduces heart neutrophil infiltration and NETs formation and ameliorates diastolic function. Lastly, SGLT2 Inhibitor empagliflozin down-regulates heart HMGB1 expression, attenuates NETs formation and cardiac fibrosis, and improves diastolic function in HFpEF mice. Conclusions NETs contribute to the pathogenesis of HFpEF, which can be ameliorated by HMGB1 inhibition and SGLT2 inhibitors. Thus, HMGB1 and NETs may represent novel therapeutic targets for the treatment of HFpEF.

Keywords

HMGB1; empagliflozin; heart failure with preserved ejection fraction; neutrophil extracellular traps.

Figures
Products