Exosomes derived from dendritic cells improve cardiac function via activation of CD4(+) T lymphocytes after myocardial infarction

J Mol Cell Cardiol. 2016 Feb:91:123-33. doi: 10.1016/j.yjmcc.2015.12.028. Epub 2015 Dec 30.

Abstract

CD4(+) T cell activation plays a key role in facilitating wound healing after myocardial infarction (MI). Exosomes (EXs) secreted from dendritic cells (DCs) can activate T cells in tumor models; however, whether DEXs (DC-EXs) can mediate CD4(+) T cell activation and improve wound healing post-MI remains unknown. This study sought to determine whether DEXs mediate CD4(+) T cell activation and improve cardiac function post-MI in mice. We used supernatants of hypoxic primary or necrotic HL-1 cardiomyocytes to simulate the post-MI cardiomyocyte microenvironment in vitro. Cultured bone marrow-derived DCs (BMDCs) from mice were stimulated with the supernatants of normal (Control group), hypoxic primary or necrotic HL-1 cardiomyocytes (MI group); a subset of BMDCs remained unstimulated (Negative group). DEXs were then isolated from the BMDC supernatants and either incubated with CD4(+) T cells or injected into mice via the tail vein. In this study, we found that the supernatants of both hypoxic primary and necrotic HL-1 cardiomyocytes upregulate DC maturation markers. After the injection of DEXs, a greater number of MI-DEXs are recruited by the mouse spleen and with greater rapidity than control- or negative-DEXs. Confocal imaging and flow cytometry revealed that MI-DEXs exhibited higher uptake by splenic CD4(+) T cells than the control- and negative-DEXs, and this increase was correlated with significantly greater increases in the expression of chemokines and the inflammatory cytokines IFN-γ and TNF by the CD4(+) T cells in vitro and in vivo. In addition, the injection of MI-DEXs improved cardiac function in mice post-MI. These results suggest that DEXs could mediate the activation of CD4(+) T cells through an endocrine mechanism and improve cardiac function post-MI. Our findings provide the basis for a novel strategy for the treatment of MI through the systemic delivery of DEXs.

Keywords: Chemokines; Cytokines; Dendritic cells; Exosomes; Myocardial infarction; T-lymphocytes.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Bone Marrow Cells / cytology
  • Bone Marrow Cells / drug effects
  • Bone Marrow Cells / immunology
  • CD4-Positive T-Lymphocytes / cytology
  • CD4-Positive T-Lymphocytes / drug effects
  • CD4-Positive T-Lymphocytes / immunology
  • Cell Hypoxia
  • Cell Movement
  • Culture Media, Conditioned / pharmacology*
  • Cytokines / biosynthesis
  • Cytokines / genetics
  • Cytokines / immunology
  • Dendritic Cells / cytology
  • Dendritic Cells / drug effects*
  • Dendritic Cells / immunology
  • Disease Models, Animal
  • Exosomes / immunology
  • Exosomes / transplantation*
  • Gene Expression Regulation
  • Lymphocyte Activation / drug effects*
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Myocardial Infarction / immunology
  • Myocardial Infarction / pathology
  • Myocardial Infarction / rehabilitation
  • Myocardial Infarction / therapy*
  • Myocardium / immunology
  • Myocardium / metabolism
  • Myocardium / pathology
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / immunology
  • Myocytes, Cardiac / pathology
  • Necrosis / immunology
  • Necrosis / pathology
  • Necrosis / rehabilitation
  • Necrosis / therapy*
  • Primary Cell Culture
  • Signal Transduction
  • Spleen / cytology
  • Spleen / drug effects
  • Spleen / immunology
  • Wound Healing / drug effects
  • Wound Healing / immunology

Substances

  • Culture Media, Conditioned
  • Cytokines