Elsevier

Cytotherapy

Volume 15, Issue 2, February 2013, Pages 140-145
Cytotherapy

Review
Mesenchymal stromal cells: misconceptions and evolving concepts

https://doi.org/10.1016/j.jcyt.2012.11.005Get rights and content

Abstract

Nearly half a century has passed since the publication of the first articles describing plastic-adherent cells from bone marrow, referred to initially as colony-forming unit fibroblasts, then marrow stromal cells, mesenchymal stem cells and most recently multipotent mesenchymal stromal cells (MSCs). As expected, our understanding of the nature and biologic functions of MSCs has undergone major paradigm shifts over this time. Despite significant advances made in deciphering their complex biology and therapeutic potential in both experimental animal models and human clinical trials, numerous misconceptions regarding the nature and function of MSCs have persisted in the field. Continued propagation of these misconceptions in some cases may significantly impede the advancement of MSC-based therapies in clinical medicine. We have identified six prevalent misconceptions about MSCs that we believe affect the field, and we attempt to rectify them based on current available data.

Introduction

Over the past several decades, concepts regarding the nature and function of mesenchymal stromal cells (MSCs) have undergone numerous major paradigm shifts. Pioneering studies by Friedenstein and colleagues first revealed that MSCs were capable of sustaining hematopoiesis and functioned as progenitors of adipogenic, chondrogenic and osteogenic lineages, properties exploited in early clinical trials 1, 2, 3. As interest in MSCs expanded, studies conducted in experimental animal models revealed the cells also possessed potent tissue reparative properties. Initial studies attributed this activity to direct cell replacement via the transdifferentiation of transplanted MSCs. However, subsequent work by many laboratories revealed that MSCs promote tissue repair via paracrine action. In recent years, the therapeutic potency of MSCs has been attributed to the secretion by cells of a large number of factors that possess angiogenic, trophic, neuro-regulatory, immunomodulatory, and anti-inflammatory activity. However, as concepts became outmoded and replaced with new paradigms, many misconceptions related to the nature and biology of MSCs arose. In this article, we identify at least six misconceptions (Figure 1) that have persisted over the years and serve as potential impediments to the successful therapeutic application of MSCs. Where possible, we attempt to clarify these misconceptions based on available published literature.

Section snippets

MSCs isolated from different tissues are equivalent

Although initially isolated from bone marrow (4) and then adipose tissue (5), MSCs or MSC-like cells have been identified in many tissues and organs. The apparent ubiquitous presence of MSCs in most tissues is attributed to their similarity to peri-vascular cells in vivo. This concept originated from studies demonstrating that bone marrow-derived MSCs express antigens common to endothelial cells and pericytes, such as STRO1 (6), CD146 and 3G5 (7), and conversely that post-capillary venule

Conclusions

The relative ease by which MSCs can be harvested and expanded to large numbers in vitro, coupled with their potent trophic, anti-inflammatory and immunomodulatory activity and lack of infusion-related toxicity in human patients has made MSCs an attractive tool for cellular therapy; this is reflected by the rapid increase in the number of ongoing MSC-based clinical trials. However, as our knowledge regarding the complex biology of MSC increases, it is necessary to discard outmoded concepts and

Acknowledgments

We would like to thank members of the MSC committee of the International Society of Cell Therapy (J. Gallipeau, M. Krampera, Y. Shi, I. Martin) and Darwin J. Prockop for their assistance with the conceptualization and development of this manuscript.

Disclosure of interest: The authors have no commercial, proprietary or financial interest in the products or companies described in this article.

References (50)

  • Y.W. Qiang et al.

    Dkk1-induced inhibitionof Wnt signaling in osteoblast differentiation is an underlying mechanism of boneloss in multiple myeloma

    Bone

    (2008)
  • E. Zappia et al.

    Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy

    Blood

    (2005)
  • G. Ren et al.

    Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide

    Cell Stem Cell

    (2008)
  • M. François et al.

    Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation

    Mol Ther

    (2012)
  • K. Le Blanc et al.

    Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study

    Lancet

    (2008)
  • D.G. Phinney

    Marrow stem

  • E.M. Horwitz et al.

    Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta

    Nat Med

    (1999)
  • O. Koc et al.

    Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy

    J Clin Oncol

    (2000)
  • A.J. Friedenstein et al.

    Fibroblast precursors in normal and irradiated mouse hematopoietic organs

    Exp Hematol

    (1976)
  • P.A. Zuk et al.

    Human adipose tissue is a source of multipotent stem cells

    Mol Biol Cell

    (2002)
  • S. Shi et al.

    Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp

    J Bone Min Res

    (2003)
  • B. Brachvogel et al.

    Perivascular cells expressing annexin A5 define a novel mesenchymal stem cell-like population with the capacity to differentiate into multiple mesenchymal lineages

    Development

    (2005)
  • L. da Silva Meirelles et al.

    Mesenchymal stem cells reside in virtually all post-natal organs and tissues

    J Cell Sci

    (2006)
  • S. Batouli et al.

    Comparison of stem-cell-mediated osteogenesis and dentinogenesis

    J Dent Res

    (2003)
  • A. Dellavalle et al.

    Pericytes resident inpostnatal skeletal muscle differentiate into muscle fibres and generate satellite cells

    Nat Comm

    (2011)
  • Cited by (0)

    View full text