Hsp22 ameliorates lipopolysaccharide-induced myocardial injury by inhibiting inflammation, oxidative stress, and apoptosis

Bioengineered. 2021 Dec;12(2):12544-12554. doi: 10.1080/21655979.2021.2010315.

Abstract

Sepsis-induced myocardial dysfunction (SIMD) is ubiquitous in septic shock patients and is associated with high morbidity and mortality rates. Heat shock protein 22 (Hsp22), which belongs to the small HSP family of proteins, is involved in several biological functions. However, the function of Hsp22 in lipopolysaccharide (LPS)-induced myocardial injury is not yet established. This study was aimed at investigating the underlying mechanistic aspects of Hsp22 in myocardial injury induced by LPS. In this study, following the random assignment of male C57BL/6 mice into control, LPS-treated, and LPS + Hsp22 treated groups, relevant echocardiograms and staining were performed to scrutinize the cardiac pathology. Plausible mechanisms were proposed based on the findings of the enzyme-linked immunosorbent assay and Western blotting assay. A protective role of Hsp22 against LPS-induced myocardial injury emerged, as evidenced from decreased levels of creatinine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and enhanced cardiac function. The post-LPS administration-caused spike in inflammatory cytokines (IL-1β, IL-6, TNF-α and NLRP3) was attenuated by the Hsp22 pre-treatment. In addition, superoxide dismutase (SOD) activity and B-cell lymphoma-2 (Bcl2) levels were augmented by Hsp22 treatment resulting in lowering of LPS-induced oxidative stress and cardiomyocyte apoptosis. In summary, the suppression of LPS-induced myocardial injury by Hsp22 overexpression via targeting of inflammation, oxidative stress, and apoptosis in cardiomyocytes paves the way for this protein to be employed in the therapy of SIMD.

Keywords: Sepsis-induced myocardial dysfunction; apoptosis; heat shock protein 22; inflammatory; lipopolysaccharide; oxidative stress.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / physiology*
  • Cardiomyopathies / chemically induced
  • Cardiomyopathies / metabolism
  • Cytokines / metabolism
  • Heat-Shock Proteins / metabolism*
  • Inflammation / chemically induced
  • Inflammation / metabolism*
  • Lipopolysaccharides / pharmacology
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Molecular Chaperones / metabolism*
  • Myocardial Ischemia / chemically induced
  • Myocardial Ischemia / metabolism*
  • Myocardium / metabolism*
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / metabolism*
  • Oxidative Stress / physiology*

Substances

  • Cytokines
  • Heat-Shock Proteins
  • Hspb8 protein, mouse
  • Lipopolysaccharides
  • Molecular Chaperones

Grants and funding

This work was supported by the National Natural Science Foundation of China (81760049, 82160070, 82100308), National Natural Science Foundation excellent youth cultivation project (20202ZDB01017), Jiangxi Science and Technology Innovation Platform Project (20165BCD41005), Natural Science Foundation of Jiangxi Province of China (20192BAB205033, 20202BABL216035), Science and technology planning project of Jiangxi Provincial Department of Education (GJJ200101);National Natural Science Foundation of China; Jiangxi Science and Technology Innovation Platform Project; Natural Science Foundation of Jiangxi Province of China;