Original article
Derivation and characterization of human fetal MSCs: An alternative cell source for large-scale production of cardioprotective microparticles

https://doi.org/10.1016/j.yjmcc.2009.12.021Get rights and content

Abstract

The therapeutic effects of mesenchymal stem cells (MSCs) transplantation are increasingly thought to be mediated by MSC secretion. We have previously demonstrated that human ESC-derived MSCs (hESC-MSCs) produce cardioprotective microparticles in pig model of myocardial ischemia/reperfusion (MI/R) injury. As the safety and availability of clinical grade human ESCs remain a concern, MSCs from fetal tissue sources were evaluated as alternatives. Here we derived five MSC cultures from limb, kidney and liver tissues of three first trimester aborted fetuses and like our previously described hESC-derived MSCs; they were highly expandable and had similar telomerase activities. Each line has the potential to generate at least 1016–19 cells or 107–10 doses of cardioprotective secretion for a pig model of MI/R injury. Unlike previously described fetal MSCs, they did not express pluripotency-associated markers such as Oct4, Nanog or Tra1-60. They displayed a typical MSC surface antigen profile and differentiated into adipocytes, osteocytes and chondrocytes in vitro. Global gene expression analysis by microarray and qRT-PCR revealed a typical MSC gene expression profile that was highly correlated among the five fetal MSC cultures and with that of hESC-MSCs (r2 > 0.90). Like hESC-MSCs, they produced secretion that was cardioprotective in a mouse model of MI/R injury. HPLC analysis of the secretion revealed the presence of a population of microparticles with a hydrodynamic radius of 50–65 nm. This purified population of microparticles was cardioprotective at ∼1/10 dosage of the crude secretion.

Introduction

Mesenchymal stem cells (MSCs) are multipotent stem cells that have a limited but robust potential to differentiate into mesenchymal cell types, e.g. adipocytes, chondrocytes and osteocytes, with negligible risk of teratoma formation. MSC transplantation has been used to treat musculoskeletal injuries, improve cardiac function in cardiovascular disease and ameliorate the severity of graft-versus-host-disease [1]. In recent years, MSC transplantations have demonstrated therapeutic efficacy in treating different diseases but the underlying mechanism has been controversial [2], [3], [4], [5], [6], [7], [8], [9]. Some reports have suggested that factors secreted by MSCs [10] were responsible for the therapeutic effect on arteriogenesis [11], stem cell crypt in the intestine [12], ischemic injury [9], [13], [14], [15], [16], [17], [18], and hematopoiesis [19], [20].

We have recently demonstrated that human MSCs derived from human embryonic stem cells (hESC-MSCs) [21] secrete > 200 proteins [22] and that a single bolus administration of hESC-MSCs conditioned medium (CM) 5 min prior to reperfusion significantly reduced infarct size by 60% and improved cardiac function in a pig and mouse model of myocardial ischemia/reperfusion (MI/R) injury [23]. In addition, this cardioprotection was mediated by large complexes of about 50–100 nm in diameter. The size of these large secreted complexes suggests that they are microparticles which are broadly defined as secreted membrane particles in the size range of 0.05–1 μm [24].

A requisite for translating cardioprotective MSC secretion into clinical applications is a clinical grade MSC source but this is currently limited by restricted access to clinical grade hESCs. Therefore, alternative tissue or cell sources that are amenable to the generation of highly expandable, clinical grade MSCs have to be developed. Here we examined fetal tissues as a candidate tissue source.

Five MSC cultures, Fllb, F1ki, F2lb, F3lb and F3li were generated from limb (lb), kidney (ki) and liver (li) tissues of three fetuses in three independent experiments. These fetal MSCs fulfilled the defining criteria of a MSC. They were highly proliferative. Each line has the potential of generating 1016–19 cells, and therefore the capacity to produce large amount of secretion. More importantly, these cells produce secretion that reduced infarct size in a mouse model of MI/R injury [23]. Like the secretion of hESC-MSCs, this cardioprotection was also mediated by large complexes. Fractionation of the secretion by size exclusion on an HPLC revealed the presence of a population of homogenously sized particles with a hydrodynamic radius of 50–65 nm and these particles were cardioprotective in a mouse model of MI/R injury. The size of these large secreted complexes suggests that they are microparticles which are broadly defined as secreted membrane particles in the size range of 0.05–1 μm [24].

Section snippets

Derivation of fetal tissue derived MSCs

The collection of fetal tissue was carried out under a KK Women's and Children's Hospital (KKH) IRB approved protocol (EC200804062) in accordance with guidelines from Singapore Bioethics Advisory Committee [25] which stated that the decision to donate the fetal tissue must be made independently from any decision to abort. Only patients who have already consented to Termination of Pregnancy (TOP) in KKH Outpatient Clinic were recruited. Recruitment was carried out in strict adherence to KKH

Generating MSC cultures from human fetal tissues

We generated five MSC cultures from fetal limb (F1lb, F2lb, F3lb), kidney (F1ki) and liver (F3li) tissues of three fetuses in three independent experiments using feeder- and serum-free culture condition as previously described [21]. A homogenous culture of putative fibroblast-like MSCs migrated out of the tissues and adhered to the plastic culture dish, 2 days after fetal tissues were plated on gelatinized tissue culture plates. This observation was consistent with the defining characteristic

Discussion

The trophic effects of MSCs transplantation on ameliorating the deleterious consequences of myocardial ischemia have been implicated in several studies [10]. Transplantation of MSCs into ischemic myocardium has been shown to induce several tissue responses such as an increased production of angiogenic factors and decreased apoptosis [38] that were better explained by secretion of paracrine factors than by differentiation of MSCs, the so-called paracrine hypothesis.

We have recently demonstrated

Disclosure statement

No potential conflict of interest.

Acknowledgments

We thank J.J. Chee (KKH) for her help in patient recruitment and tissue harvest, and Jayanthi Padmanabhan (BTI) for technical assistance in preparing the secretion.

References (59)

  • A. Aharon et al.

    Microparticles, thrombosis and cancer

    Best Pract. Res. Clin. Haematol.

    (2009)
  • C.T. Esmon

    Basic mechanisms and pathogenesis of venous thrombosis

    Blood Rev.

    (2009)
  • E. Stein et al.

    The coagulopathy of acute promyelocytic leukaemia revisited

    Best Pract. Res. Clin. Haematol.

    (2009)
  • K. Hartvigsen et al.

    The role of innate immunity in atherogenesis

    J. Lipid Res.

    (2009)
  • T. Benameur et al.

    Therapeutic potential of plasma membrane-derived microparticles

    Pharmacol. Rep.

    (2009)
  • J.J. Minguell et al.

    Mesenchymal stem cells and the treatment of cardiac disease

    Exp. Biol. Med. (Maywood)

    (2006)
  • K.H. Schuleri et al.

    Mesenchymal stem cells for cardiac regenerative therapy

    Handb. Exp. Pharmacol.

    (2007)
  • A. Abdel-Latif et al.

    Adult bone marrow-derived cells for cardiac repair: a systematic review and meta-analysis

    Arch. Intern. Med.

    (2007)
  • S. Ohnishi et al.

    Prepare cells to repair the heart: mesenchymal stem cells for the treatment of heart failure

    Am. J. Nephrol.

    (2007)
  • D.E. Atsma et al.

    Opportunities and challenges for mesenchymal stem cell-mediated heart repair

    Curr. Opin. Lipidol.

    (2007)
  • M. Gnecchi et al.

    Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells

    Nat. Med.

    (2005)
  • A.I. Caplan et al.

    Mesenchymal stem cells as trophic mediators

    J. Cell Biochem.

    (2006)
  • T. Kinnaird et al.

    Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms

    Circulation

    (2004)
  • S.J. Leedham et al.

    Intestinal stem cells

    J Cell Mol Med

    (2005)
  • F. Togel et al.

    Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms

    Am. J. Physiol. Renal Physiol.

    (2005)
  • Y. Miyahara et al.

    Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction

    Nat. Med.

    (2006)
  • M. Gnecchi et al.

    Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement

    FASEB J.

    (2006)
  • H. Mayer et al.

    Vascular endothelial growth factor (VEGF-A) expression in human mesenchymal stem cells: autocrine and paracrine role on osteoblastic and endothelial differentiation

    J. Cell Biochem.

    (2005)
  • H. Nakagami et al.

    Novel autologous cell therapy in ischemic limb disease through growth factor secretion by cultured adipose tissue-derived stromal cells

    Arterioscler. Thromb. Vasc. Biol.

    (2005)
  • Cited by (116)

    • Engineered exosomes for cancer theranostics: Next-generation tumor targeting

      2023, Journal of Drug Delivery Science and Technology
    • Therapeutic effect of co-culture of rat bone marrow mesenchymal stem cells and degenerated nucleus pulposus cells on intervertebral disc degeneration

      2021, Spine Journal
      Citation Excerpt :

      With expanding research into this subject, it was found that biological information can be transferred between cells that are not in contact through paracrine mechanisms or exosomes [9]. Yang [10], Yammamoto [11], and others pointed out that under physiological conditions, the interaction between BMSCs and NPCs is a non-contact method. In 2010, Lai et al. [12] found that MSCs can secrete exosomes, which are class-specific extracellular vesicles with a diameter of 40 to 100 nm.

    View all citing articles on Scopus
    View full text