Review
Circulating mesenchymal stem cells

https://doi.org/10.1016/j.biocel.2003.10.007Get rights and content

Abstract

Mesenchymal precursor cells (MPCs) are multipotent cells capable of differentiating into various mesenchymal tissues, such as bone, cartilage, fat, tendon and muscle. They are present within both mesenchymal tissues and the bone marrow (BM). If marrow-derived MPCs are to have a role in repair and fibrosis of mesenchymal tissues, transit of these cells through the peripheral blood is to be expected. Although there is evidence for the existence of MPCs within the peripheral blood, results are debated and are not always reproducible. Variations in the methods of cell purification, culture and characterisation may explain the inconsistent results obtained in different studies.

Introduction

The study of adult type stem cells is a hot subject, because of the insights they offer into the understanding of tissue repair and regeneration, and also because they represent a possible alternative to embryonic stem (ES) cells in various therapeutic applications. Although haematopoietic stem cells are the best characterised, adult organisms contain several other types such as neural, epithelial and mesenchymal stem cells (MSCs). As will be discussed below, MSCs are found both in mesenchymal tissues and within the bone marrow (BM). Recent investigations have shown that after systemic infusion, marrow-derived MSCs engraft within multiple tissues of mesenchymal origin in the adult organism (Pereira et al., 1995, Prockop, 1997). How bone marrow-derived MSCs gain access to mesenchymal tissues, and whether they play a role in physiological turnover of these tissues, remains unknown. Transit of the MSCs through the peripheral blood would be expected to bridge the gap between the bone marrow and the mesenchymal tissues in need of repair. This article reviews current data on the existence of such a population of circulating MSCs.

Section snippets

What are mesenchymal stem cells?

Excellent reviews of mesenchymal stem cells have already been published (Minguell, Erices, & Conget, 2001), but a brief summary will be provided here.

“Mesenchyme” designates the developing loose connective tissue of an embryo, mainly derived from the mesoderm, and giving rise to a large part of the cells of the connective tissue in the adult. The definition is generally extended to include connective tissue cells in adult tissues such as (myo)fibroblasts, bone, cartilage, fat, tendon, muscles,

Do mesenchymal progenitor cells circulate in the peripheral blood?

Several studies address this issue by attempting to isolate MPCs from peripheral blood using culture conditions similar to those defined for bone marrow-derived MPCs, in either adult or fetal organisms. These studies, which show conflicting results, will be described first.

Other studies have examined the fate of marrow-derived cells after systemic infusion. The engraftment of these cells in various mesenchymal tissues provides indirect evidence for circulating MPCs. Similarly, so does the

Transplantation studies

As already mentioned above, in vivo experiments involving bone marrow transplantation and solid organ transplantation can provide indirect evidence for the existence of circulating MPCs.

Circulating BM-derived endothelial precursor cells (hemangioblasts)

If there is good evidence that bone marrow-derived cells can circulate and seed many tissues and organs, what of the possibility that these same, or similar, cells can also seed new or existing endothelium? The answer, broadly, is yes, and recent evidence is becoming stronger that this is a frequent event under certain circumstances. For example, it has been shown in vitro that cells derived from fetal bone marrow can differentiate into endothelial and haemopoietic cells (Guo et al., 2003). In

Do circulating MAPCs exist?

A subset of marrow cells that co-purify with MPCs, termed multipotent adult progenitor cells (MAPCs) appear to have considerable plasticity, in that, in addition to generating mesenchymal cell types, they are also able to give rise to endothelial cells, neural cells and epithelial cells in vitro under defined culture conditions (Jiang et al., 2002a, Reyes et al., 2001). MAPCs have been purified from human, murine or rat bone marrow mononuclear cells. In vivo after injection into blastocysts,

Conclusions

Mesenchymal stem cells have the potential to be a source of multipotent cells for autologous cell and gene therapy. Following isolation, purification and possibly in vitro differentiation, MPCs could be injected for local cell therapy in patients with damaged bone, cartilage or tendon (Minguell et al., 2001). Genetically modified MPCs could also be used to replace dysfunctional mesenchymal cells, as suggested in the study by Horwitz et al. mentioned earlier (Horwitz et al., 1999). Adult bone

Acknowledgements

Dr. Candice Roufosse is supported by a Clinical Research Fellowship Grant from the Wellcome Trust.

References (71)

  • Y. Jiang et al.

    Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain

    Experimental Hematology

    (2002)
  • E.L. Lagaaij et al.

    Endothelial cell chimerism after renal transplantation and vascular rejection

    Lancet

    (2001)
  • I. Moreau et al.

    Myofibroblastic stromal cells isolated from human bone marrow induce the proliferation of both early myeloid and B-lymphoid cells

    Blood

    (1993)
  • M. Reyes et al.

    Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells

    Blood

    (2001)
  • P.J. Simmons et al.

    Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1

    Blood

    (1991)
  • C.M. Verfaillie

    Adult stem cells: Assessing the case for pluripotency

    Trends in Cell Biology

    (2002)
  • L. Yang et al.

    Peripheral blood fibrocytes from burn patients: Identification and quantification of fibrocytes in adherent cells cultured from peripheral blood mononuclear cells

    Laboratory Investigation

    (2002)
  • M.R. Alison et al.

    Plastic adult stem cells: Will they graduate from the school of hard knocks?

    Journal of Cell Science

    (2003)
  • P. Anklesaria et al.

    Engraftment of a clonal bone marrow stromal cell line in vivo stimulates hematopoietic recovery from total body irradiation

    Proceedings of the National Academic Science of United States of America

    (1987)
  • E.V. Badiavas et al.

    Participation of bone marrow derived cells in cutaneous wound healing

    Journal of Cell Physiology

    (2003)
  • Bailey, A. S., Jiang, S., Afentoulis, M., Baumann, C. I., Schroeder, D. A., Olson, S. B., Wong, M. H., & Fleming, W. H....
  • M. Brittan et al.

    Bone marrow derivation of pericryptal myofibroblasts in the mouse and human small intestine and colon

    Gut

    (2002)
  • S.P. Bruder et al.

    Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation

    Journal of Cell Biochemistry

    (1997)
  • R. Bucala et al.

    Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair

    Molecular Medicine

    (1994)
  • Cherry, Yasumizu, R., Toki, J., Asou, H., Nishino, T., Komatsu, Y., & Ikehara, S. (1994). Production of hematopoietic...
  • J. Chesney et al.

    Regulated production of type I collagen and inflammatory cytokines by peripheral blood fibrocytes

    Journal of Immunology

    (1998)
  • Cogle, C. R., Wainman, D. A., Jorgensen, M. L., Guthrie, S. M., Mames, R. N., & Scott, E. W. (2004). Adult human...
  • M.C. del Canizo et al.

    Hematopoietic damage prior to PBSCT and its influence on hematopoietic recovery

    Haematologica

    (1999)
  • C.M. Digirolamo et al.

    Propagation and senescence of human marrow stromal cells in culture: A simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate

    British Journal of Haematology

    (1999)
  • Direkze, N. C., Forbes, S. J., Brittan, M., Hunt, T., Jeffery, R., Preston, S. L., Poulsom, R., Hodivala-Dilke, K.,...
  • A. Eichmann et al.

    Vasculogenesis and the search for the hemangioblast

    Journal of Hematotherpy and Stem Cell Research

    (2002)
  • M.W. Epperly et al.

    Bone marrow origin of myofibroblasts in irradiation pulmonary fibrosis

    American Journal of Respiraory Cell and Molecular Biology

    (2003)
  • A. Erices et al.

    Mesenchymal progenitor cells in human umbilical cord blood

    British Journal of Haematology

    (2000)
  • M. Fernandez et al.

    Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients

    Bone Marrow Transplantation

    (1997)
  • G. Ferrari et al.

    Muscle regeneration by bone marrow-derived myogenic progenitors

    Science

    (1998)
  • Cited by (263)

    • Comparison of Bone Formation After Sinus Membrane Lifting Without Graft or Using Bone Substitute “Histologic and Radiographic Evaluation”

      2021, Journal of Oral and Maxillofacial Surgery
      Citation Excerpt :

      In addition, the clot can be a scaffold, creating a center for the initiation of new bone formation. As reported before, mesenchymal stem cells (MSCs) derived from peripheral blood may contribute to the bone formation in the sinus compartment.27 These cells, routinely present in the peripheral circulating blood in low quantities but can be mobilized in large numbers to tissues undergoing traumatic events.

    • Microfluidic devices for stem cell analysis

      2021, Microfluidic Devices for Biomedical Applications
    View all citing articles on Scopus
    View full text