Migration towards SDF-1 selects angiogenin-expressing bone marrow monocytes endowed with cardiac reparative activity in patients with previous myocardial infarction

Stem Cell Res Ther. 2015 Apr 11;6(1):53. doi: 10.1186/s13287-015-0028-y.

Abstract

Introduction: Chemokine-directed migration is crucial for homing of regenerative cells to the infarcted heart and correlates with outcomes of cell therapy trials. Hence, transplantation of chemokine-responsive bone marrow cells may be ideal for treatment of myocardial ischemia. To verify the therapeutic activity of bone marrow mononuclear cells (BM-MNCs) selected by in vitro migration towards the chemokine stromal cell-derived factor-1 (SDF-1) in a mouse model of myocardial infarction (MI), we used BM-MNCs from patients with previous large MI recruited in the TransACT-1&2 cell therapy trials.

Methods: Unfractioned BM-MNCs, SDF-1-responsive, and SDF-1-nonresponsive BM-MNCs isolated by patients recruited in the TransACT-1&2 cell therapy trials were tested in Matrigel assay to evaluate angiogenic potential. Secretome and antigenic profile were characterized by flow cytometry. Angiogenin expression was measured by RT-PCR. Cells groups were also intramyocardially injected in an in vivo model of MI (8-week-old immune deficient CD1-FOXN1(nu/nu) mice). Echocardiography and hemodynamic measurements were performed before and at 14 days post-MI. Arterioles and capillaries density, infiltration of inflammatory cells, interstitial fibrosis, and cardiomyocyte proliferation and apoptosis were assessed by immunohistochemistry.

Results: In vitro migration enriched for monocytes, while CD34(+) and CD133(+) cells and T lymphocytes remained mainly confined in the non-migrated fraction. Unfractioned total BM-MNCs promoted angiogenesis on Matrigel more efficiently than migrated or non-migrated cells. In mice with induced MI, intramyocardial injection of unfractionated or migrated BM-MNCs was more effective in preserving cardiac contractility and pressure indexes than vehicle or non-migrated BM-MNCs. Moreover, unfractioned BM-MNCs enhanced neovascularization, whereas the migrated fraction was unique in reducing the infarct size and interstitial fibrosis. In vitro studies on isolated cardiomyocytes suggest participation of angiogenin, a secreted ribonuclease that inhibits protein translation under stress conditions, in promotion of cardiomyocyte survival by migrated BM-MNCs.

Conclusions: Transplantation of bone marrow cells helps post-MI healing through distinct actions on vascular cells and cardiomyocytes. In addition, the SDF-1-responsive fraction is enriched with angiogenin-expressing monocytes, which may improve cardiac recovery through activation of cardiomyocyte response to stress. Identification of factors linking migratory and therapeutic outcomes could help refine regenerative approaches.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • AC133 Antigen
  • Animals
  • Antigens, CD / metabolism
  • Antigens, CD34 / metabolism
  • Bone Marrow Cells / cytology
  • Bone Marrow Cells / drug effects*
  • Cell Movement / drug effects
  • Chemokine CXCL12 / pharmacology*
  • Cytokines / analysis
  • Disease Models, Animal
  • Echocardiography
  • Glycoproteins / metabolism
  • Hemodynamics
  • Humans
  • Intercellular Signaling Peptides and Proteins / analysis
  • Male
  • Mice
  • Middle Aged
  • Monocytes / cytology
  • Monocytes / metabolism
  • Monocytes / transplantation*
  • Myocardial Ischemia / pathology
  • Myocardial Ischemia / therapy*
  • Myocytes, Cardiac / cytology
  • Myocytes, Cardiac / metabolism
  • Peptides / metabolism
  • Ribonuclease, Pancreatic / genetics
  • Ribonuclease, Pancreatic / metabolism*

Substances

  • AC133 Antigen
  • Antigens, CD
  • Antigens, CD34
  • Chemokine CXCL12
  • Cytokines
  • Glycoproteins
  • Intercellular Signaling Peptides and Proteins
  • PROM1 protein, human
  • Peptides
  • Prom1 protein, mouse
  • angiogenin
  • Ribonuclease, Pancreatic