Sectm1a deficiency aggravates inflammation-triggered cardiac dysfunction through disruption of LXRα signalling in macrophages

Cardiovasc Res. 2021 Feb 22;117(3):890-902. doi: 10.1093/cvr/cvaa067.

Abstract

Aims: Cardiac dysfunction is a prevalent comorbidity of disrupted inflammatory homeostasis observed in conditions such as sepsis (acute) or obesity (chronic). Secreted and transmembrane protein 1a (Sectm1a) has previously been implicated to regulate inflammatory responses, yet its role in inflammation-associated cardiac dysfunction is virtually unknown.

Methods and results: Using the CRISPR/Cas9 system, we generated a global Sectm1a-knockout (KO) mouse model and observed significantly increased mortality and cardiac injury after lipopolysaccharide (LPS) injection, when compared with wild-type (WT) control. Further analysis revealed significantly increased accumulation of inflammatory macrophages in hearts of LPS-treated KO mice. Accordingly, ablation of Sectm1a remarkably increased inflammatory cytokines levels both in vitro [from bone marrow-derived macrophages (BMDMs)] and in vivo (in serum and myocardium) after LPS challenge. RNA-sequencing results and bioinformatics analyses showed that the most significantly down-regulated genes in KO-BMDMs were modulated by LXRα, a nuclear receptor with robust anti-inflammatory activity in macrophages. Indeed, we identified that the nuclear translocation of LXRα was disrupted in KO-BMDMs when treated with GW3965 (LXR agonist), resulting in higher levels of inflammatory cytokines, compared to GW3965-treated WT-cells. Furthermore, using chronic inflammation model of high-fat diet (HFD) feeding, we observed that infiltration of inflammatory monocytes/macrophages into KO-hearts were greatly increased and accordingly, worsened cardiac function, compared to WT-HFD controls.

Conclusion: This study defines Sectm1a as a new regulator of inflammatory-induced cardiac dysfunction through modulation of LXRα signalling in macrophages. Our data suggest that augmenting Sectm1a activity may be a potential therapeutic approach to resolve inflammation and associated cardiac dysfunction.

Keywords: Cardiac function; Cardiac inflammation; Inflammation; LXR; Macrophage.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cytokines / genetics
  • Cytokines / metabolism
  • Diet, High-Fat
  • Disease Models, Animal
  • Gene Expression Regulation
  • Heart Diseases / etiology
  • Heart Diseases / genetics
  • Heart Diseases / metabolism*
  • Heart Diseases / physiopathology
  • Inflammation / etiology
  • Inflammation / genetics
  • Inflammation / metabolism*
  • Inflammation / physiopathology
  • Inflammation Mediators / metabolism
  • Lipopolysaccharides
  • Liver X Receptors / genetics
  • Liver X Receptors / metabolism*
  • Macrophages / metabolism*
  • Male
  • Membrane Proteins / deficiency*
  • Membrane Proteins / genetics
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Phenotype
  • RAW 264.7 Cells
  • Rats
  • Rats, Sprague-Dawley
  • Signal Transduction
  • Ventricular Function, Left*

Substances

  • Cytokines
  • Inflammation Mediators
  • Lipopolysaccharides
  • Liver X Receptors
  • Membrane Proteins
  • Nr1h3 protein, mouse
  • SECTM1A protein, mouse