Anti-CD3 Antibody Treatment Reduces Scar Formation in a Rat Model of Myocardial Infarction

Cells. 2020 Jan 25;9(2):295. doi: 10.3390/cells9020295.

Abstract

: Introduction: Antibody treatment with anti-thymocyte globulin (ATG) has been shown to be cardioprotective. We aimed to evaluate which single anti-T-cell epitope antibody alters chemokine expression at a level similar to ATG and identified CD3, which is a T-cell co-receptor mediating T-cell activation. Based on these results, the effects of anti-CD3 antibody treatment on angiogenesis and cardioprotection were tested in vitro and in vivo.

Methods: Concentrations of IL-8 and MCP-1 in supernatants of human peripheral blood mononuclear cell (PBMC) cultures following distinct antibody treatments were evaluated by Enzyme-linked Immunosorbent Assay (ELISA). In vivo, anti-CD3 antibodies or vehicle were injected intravenously in rats subjected to acute myocardial infarction (AMI). Chemotaxis and angiogenesis were evaluated using tube and migration assays. Intracellular pathways were assessed using Western blot. Extracellular vesicles (EVs) were quantitatively evaluated using fluorescence-activated cell scanning, exoELISA, and nanoparticle tracking analysis. Also, microRNA profiles were determined by next-generation sequencing.

Results: Only PBMC stimulation with anti-CD3 antibody led to IL-8 and MCP-1 changes in secretion, similar to ATG. In a rat model of AMI, systemic treatment with an anti-CD3 antibody markedly reduced infarct scar size (27.8% (Inter-quartile range; IQR 16.2-34.9) vs. 12.6% (IQR 8.3-27.2); p < 0.01). The secretomes of anti-CD3 treated PBMC neither induced cardioprotective pathways in cardiomyocytes nor pro-angiogenic mechanisms in human umbilical vein endothelial cell (HUVECs) in vitro. While EVs quantities remained unchanged, PBMC incubation with an anti-CD3 antibody led to alterations in EVs miRNA expression.

Conclusion: Treatment with an anti-CD3 antibody led to decreased scar size in a rat model of AMI. Whereas cardioprotective and pro-angiogenetic pathways were unaltered by anti-CD3 treatment, qualitative changes in the EVs miRNA expression could be observed, which might be causal for the observed cardioprotective phenotype. We provide evidence that EVs are a potential cardioprotective treatment target. Our findings will also provide the basis for a more detailed analysis of putatively relevant miRNA candidates.

Keywords: AMI; CD3; angiogenesis; anti-CD; antibody treatment; apoptosis; cardioprotection; miRNA; myocardial infarction.

MeSH terms

  • Animals
  • Antibodies / immunology
  • Antibodies / therapeutic use
  • Antilymphocyte Serum / immunology
  • Antilymphocyte Serum / therapeutic use
  • CD3 Complex / immunology*
  • Cardiotonic Agents / immunology
  • Chemokine CCL2 / metabolism
  • Cicatrix / drug therapy*
  • Cicatrix / immunology
  • Cicatrix / prevention & control
  • Disease Models, Animal
  • Exosomes / drug effects
  • Exosomes / immunology
  • Exosomes / metabolism
  • Extracellular Vesicles / drug effects
  • Extracellular Vesicles / metabolism
  • High-Throughput Nucleotide Sequencing
  • Human Umbilical Vein Endothelial Cells
  • Humans
  • Interleukin-8 / metabolism
  • Leukocytes, Mononuclear / drug effects*
  • Leukocytes, Mononuclear / immunology
  • Leukocytes, Mononuclear / metabolism
  • Male
  • MicroRNAs / genetics
  • MicroRNAs / metabolism*
  • Myocardial Infarction / drug therapy*
  • Myocardial Infarction / immunology*
  • Neovascularization, Physiologic / drug effects*
  • Neovascularization, Physiologic / immunology
  • Proteome / metabolism
  • Rats
  • Rats, Sprague-Dawley

Substances

  • Antibodies
  • Antilymphocyte Serum
  • CCL2 protein, human
  • CD3 Complex
  • Cardiotonic Agents
  • Chemokine CCL2
  • Interleukin-8
  • MicroRNAs
  • Proteome