Splenic Ly6Chi monocytes contribute to adverse late post-ischemic left ventricular remodeling in heme oxygenase-1 deficient mice

Basic Res Cardiol. 2017 Jul;112(4):39. doi: 10.1007/s00395-017-0629-y. Epub 2017 May 22.

Abstract

Heme oxygenase-1 (Hmox1) is a stress-inducible protein crucial in heme catabolism. The end products of its enzymatic activity possess anti-oxidative, anti-apoptotic and anti-inflammatory properties. Cardioprotective effects of Hmox1 were demonstrated in experimental models of myocardial infarction (MI). Nevertheless, its importance in timely resolution of post-ischemic inflammation remains incompletely understood. The aim of this study was to determine the role of Hmox1 in the monocyte/macrophage-mediated cardiac remodeling in a mouse model of MI. Hmox1 knockout (Hmox1-/-) and wild-type (WT, Hmox1+/+) mice were subjected to a permanent ligation of the left anterior descending coronary artery. Significantly lower incidence of left ventricle (LV) free wall rupture was noted between 3rd and 5th day after MI in Hmox1-/- mice resulting in their better overall survival. Then, starting from 7th until 21st day post-MI a more potent deterioration of LV function was observed in Hmox1-/- than in the surviving Hmox1+/+ mice. This was accompanied by higher numbers of Ly6Chi monocytes in peripheral blood, as well as higher expression of monocyte chemoattractant protein-1 and adhesion molecules in the hearts of MI-operated Hmox1-/- mice. Consequently, a greater post-MI monocyte-derived myocardial macrophage infiltration was noted in Hmox1-deficient individuals. Splenectomy decreased the numbers of circulating inflammatory Ly6Chi monocytes in blood, reduced the numbers of proinflammatory cardiac macrophages and significantly improved the post-MI LV function in Hmox1-/- mice. In conclusion, Hmox1 deficiency has divergent consequences in MI. On the one hand, it improves early post-MI survival by decreasing the occurrence of cardiac rupture. Afterwards, however, the hearts of Hmox1-deficient mice undergo adverse late LV remodeling due to overactive and prolonged post-ischemic inflammatory response. We identified spleen as an important source of these cardiovascular complications in Hmox1-/- mice.

Keywords: Cardiac rupture; Heme oxygenase-1; Macrophages; Monocytes; Myocardial infarction.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antigens, Ly / immunology
  • Antigens, Ly / metabolism*
  • Bone Marrow Cells / enzymology
  • Disease Models, Animal
  • Female
  • Genotype
  • Heart Rupture, Post-Infarction / enzymology
  • Heart Rupture, Post-Infarction / pathology
  • Heart Rupture, Post-Infarction / physiopathology
  • Hematopoiesis
  • Heme Oxygenase-1 / deficiency*
  • Heme Oxygenase-1 / genetics
  • Macrophages / enzymology
  • Macrophages / immunology
  • Membrane Proteins / deficiency*
  • Membrane Proteins / genetics
  • Mice, Knockout
  • Monocytes / enzymology*
  • Monocytes / immunology
  • Myocardial Infarction / enzymology*
  • Myocardial Infarction / immunology
  • Myocardial Infarction / pathology
  • Myocardial Infarction / physiopathology
  • Myocardium / enzymology*
  • Myocardium / pathology
  • Phenotype
  • Spleen / enzymology*
  • Spleen / immunology
  • Time Factors
  • Ventricular Dysfunction, Left / enzymology
  • Ventricular Dysfunction, Left / pathology
  • Ventricular Dysfunction, Left / physiopathology
  • Ventricular Function, Left*
  • Ventricular Remodeling*

Substances

  • Antigens, Ly
  • Ly-6C antigen, mouse
  • Membrane Proteins
  • Heme Oxygenase-1
  • Hmox1 protein, mouse