Elsevier

Life Sciences

Volume 80, Issue 6, 16 January 2007, Pages 559-565
Life Sciences

Transplantation of adipose stromal cells, but not mature adipocytes, augments ischemia-induced angiogenesis

https://doi.org/10.1016/j.lfs.2006.10.020Get rights and content

Abstract

Therapeutic angiogenesis has emerged as a promising therapy to treat patients with ischemic diseases. Transplantation of bone marrow cells (BMCs) is reported to augment collateral development in ischemic organs either by differentiating into vascular cells or by secreting angiogenic cytokines. Recent evidence suggests that adipose tissues secrete a number of humoral factors and contain pluripotent stem cells. Here, we evaluated the therapeutic potential of adipose tissue-derived cells to promote angiogenesis in a mouse model of hind limb ischemia. Stromal vascular fraction cells (SVFs) were isolated from inguinal adipose tissue. Endothelial-like cells or smooth muscle-like cells could be obtained from the culture of SVFs in the presence of growth factors. Freshly isolated BMCs, SVFs, or mature adipocytes were transplanted into the ischemic hind limb of mice. SVFs significantly augmented collateral development as determined by the restoration of blood perfusion and capillary density of the ischemic muscle. Angiogenic effects of SVFs were as potent as those of BMCs. Mature adipocytes showed no proangiogenic effects. The ischemic muscle contained endothelial cells or smooth muscle cells that derived from the transplanted SVFs and BMCs. These results suggest that SVFs might be used to promote angiogenesis in ischemic tissues.

Introduction

Angiogenesis is a physiological response to ischemia (Folkman, 1995). In animal models of ischemia, a large body of evidence indicates that administration of angiogenic growth factors can augment nutrients perfusion through neovascularization (Freedman and Isner, 2001). Therapeutic angiogenesis, a strategy to cure tissue ischemia by promoting collateral growth, has emerged as one of the most promising therapies to date for ischemic diseases (Freedman and Isner, 2001). Although early clinical trials reported that the administration of angiogenic growth factors as a recombinant protein or gene could enhance the formation of new collateral vessels, relieving some ischemic symptoms (Freedman and Isner, 2001), the clinical efficacy of administration of a single growth factor is still controversial (Henry et al., 2003, Kastrup et al., 2005).

Accumulating evidence suggests that adult body may contain multipotent stem cells and/or progenitors of vascular endothelial cells and smooth muscle cells (Asahara et al., 1997, Sata, 2006, Simper et al., 2002). Animal and clinical studies demonstrated that transplanted bone marrow cells or endothelial progenitor cells promoted collateral formation in ischemic tissues by differentiating into vascular cells and/or by secreting angiogenic cytokines (Kamihata et al., 2001, Tateishi-Yuyama et al., 2002, Urbich and Dimmeler, 2004). Double blinded clinical studies are being performed to test the efficacy and the safety of autologous transplantation of bone marrow cells or ex vivo expanded autologous endothelial cells in patients with acute myocardial infarction (Drexler et al., 2006, Wollert et al., 2004). However, bone marrow aspiration is an invasive procedure that can not be performed frequently. Although endothelial progenitor cells could be mobilized into the systemic circulation by cytokines (Takahashi et al., 1999), the safety of the procedure is still controversial (Kang et al., 2004). Therefore, alternative source of stem cells or progenitors for therapeutic angiogenesis has been desired.

Recently, adipose tissues are reported to contain multipotent mesenchymal stem cells that have the ability to regenerate damaged organs (Gimble and Guilak, 2003, Miranville et al., 2004, Miyahara et al., 2006, Planat-Benard et al., 2004, Zuk et al., 2002). Here, we investigated therapeutic efficacy of stromal vascular fraction cells (SVFs) or mature adipocytes freshly isolated from adipose tissues in a murine model of hind limb ischemia. Results suggest that adipose-derived SVFs, but not mature adipocytes, are as potent as bone marrow cells in promoting angiogenesis when transplanted into ischemic tissues.

Section snippets

Animals

C57BL/6 mice were purchased from SLC (Shizuoka, Japan). Transgenic mice (C57BL/6 background) that ubiquitously express enhanced green fluorescent protein (GFP mice) were a generous gift from Dr Masaru Okabe (Osaka University, Osaka, Japan) (Okabe et al., 1997). All mice were kept in microisolator cages on a 12-h day/night cycle. All experimental procedures and protocols were approved by the Animal Care and Use Committee of the University of Tokyo and complied with the “Guide for the Care and

Endothelial-like cells and smooth muscle-like cells obtained from SVFs in vitro

SVFs were plated on a fibronectin-coated plate and cultured in EBM supplemented with bovine brain extract, vascular endothelial growth factor, and 20% FBS (endothelial progenitor cell medium). At 14 days, there were adherent cells that were doubly stained by DiI-Ac-LDL and FITC-BS lectin (Fig. 1A). SVFs were also cultured in the presence of PDGF-BB and bFGF. When the medium was changed at 4 days, there were adherent cells that consisted of heterogeneous cell types. Adherent cells showed a

Discussion

In this study, we found that endothelial-like cells and smooth muscle-like cells could be obtained from the culture of SVFs isolated from adipose tissues. SVFs, but not mature adipocytes, augmented new vessel formation when transplanted into the ischemic hind limb. The angiogenic effect of SVFs was as potent as that of BMCs. At three weeks, there were endothelial cells and smooth muscle cells that derived from the transplanted SVFs and BMCs.

Adipose tissue is mainly composed of two different

Acknowledgements

This study was supported in part by grants from the Ministry of Education, Culture, Sports, Science and Technology and the Ministry of Health, Labor and Welfare of Japan.

References (34)

  • H. Drexler et al.

    Bone-marrow-derived cell transfer after st-elevation myocardial infarction: lessons from the boost trial

    Nature Clinical Practice. Cardiovascular Medicine

    (2006)
  • J. Folkman

    Seminars in medicine of the Beth Israel hospital, Boston. Clinical applications of research on angiogenesis

    New England Journal of Medicine

    (1995)
  • S. Freedman et al.

    Therapeutic angiogenesis for ischemic cardiovascular disease

    Journal of Molecular and Cellular Cardiology

    (2001)
  • D. Fukuda et al.

    Potent inhibitory effect of sirolimus on circulating vascular progenitor cells

    Circulation

    (2005)
  • D. Fukumura et al.

    Paracrine regulation of angiogenesis and adipocyte differentiation during in vivo adipogenesis

    Circulation Research

    (2003)
  • T.D. Henry et al.

    The viva trial: vascular endothelial growth factor in ischemia for vascular angiogenesis

    Circulation

    (2003)
  • H. Kamihata et al.

    Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines

    Circulation

    (2001)
  • Cited by (0)

    View full text