Elsevier

Ageing Research Reviews

Volume 5, Issue 1, February 2006, Pages 91-116
Ageing Research Reviews

Review
Aging of mesenchymal stem cells

https://doi.org/10.1016/j.arr.2005.10.001Get rights and content

Abstract

The role of adult mesenchymal stem cells (MSC) in tissue maintenance and regeneration has received significant attention of late. Questions arise to what extent these cells are either subject to, or causes of aging; whether age-related changes in these cells are due to intrinsic factors or induced by the somatic environment.

This review collates and examines recent data in support of these different theories.

By means of introduction, a brief overview is given of current MSC definitions and their basic role in tissue regeneration followed by a comparative analysis of gerontological studies involving MSC. Evidence for extrinsic aging and various aging markers relating to morphology, proliferation, signalling, senescence markers, telomeres and telomerase, and other indicators is discussed.

We observe that while the literature might often appear to conflict, many apparent discrepancies are attributable to inconsistent methods of extracting and isolating MSC which in fact contains various subsets of adult stem cells, varying not only in their differentiation potential but also in their vulnerability to senescence—ranging from quasi-somatic lifespan to perennial vigour.

Thus, mesenchymal stem cells emerge as both subject to and key mediators of organismal aging.

Introduction

In recent years, multipotent stem cells in adult tissue have received considerable attention. The term mesenchymal stem cells (MSC) was popularized by Caplan (Gao et al., 2001), in reference to work by Friedenstein and Owen (Friedenstein et al., 1970) describing a plastic-adherent fibroblastic cell isolated by Percoll density centrifugation, reactive with monoclonal antibodies SH2 and SH3.

The adjective ‘mesenchymal’ is fraught with some ambiguity since ‘mesenchyme’ describes tissue of mesodermal origin, the middle embryological germ layer, giving rise to the musculoskeletal, blood, vascular and urinogenital systems, and to connective tissue (including dermis). Thus, developmentally speaking, the term ‘mesenchymal’ should include both blood and connective tissue cells.

In practice however, only the latter cells are usually described as being derived from MSC and considered distinct from haematopoietic stem cells (HSC), which are responsible for the development, maintenance, and regeneration of blood forming tissues [reviewed by Chen (2002)]. It is quite possible that MSC and HSC have a common precursor in the elusive “haemangioblasts” (Sabin, 1920), in the cells identified by the group of Verfaillie originally termed “mesodermal progenitor” (Reyes et al., 2001), later “multipotent adult progenitor” (MAPC) (Young and Black, 2004) cells, or in “pluripotent stem cells” (Howell et al., 2003), or ‘tissue committed stem cells’ (TCSC) (Ratajczak et al., 2004). However, this is contentious and the physiological relevance of these cells remains to be demonstrated.

Cells with non-haematopoietic multipotency can ultimately differentiate into multiple cell lineages including osteoblasts (Jaiswal et al., 1997), adipocytes (Purpura et al., 2004), chondrocytes (Johnstone et al., 1998), myoblasts (Wakitani et al., 1995), and early progenitors of neural cells (Deng et al., 2001). Such cells can be isolated from umbilical cord blood (Lee et al., 2004, Hou et al., 2003), connective tissue (Young et al., 1995), skin (Shih et al., 2005), synovium fluid (Jones et al., 2004), fat (De Ugarte et al., 2003), the placenta (Waller et al., 1995) and even teeth (Nakashima and de Crombrugghe, 2003), but most commonly they are taken from marrow of various bones.

Accordingly, the nomenclature is not consistent. Designations for cells with non-haematopoietic multipotency have included “colony-forming-unit-fibroblasts”, “stromal (stem) cells”, “bone marrow (stromal) cells”, “skeletal stem cells”, “mesodermal progenitor cells”, “non-haematopoietic stem cells”, “(bone marrow) stem cells”, “mesenchymal progenitor cells” and others (see Baksh et al., 2004, Young and Black, 2004 for enumeration). There is also an understandable tendency to designate such cells as “pre-(lineage-under-investigation)” cells (e.g. pre-osteoblast etc.). It has also been suggested that MSC are simply pericytes (Nakashima and de Crombrugghe, 2003).

Some of the inconsistencies surrounding the identification of MSC arise from the fact that specific markers have not yet been agreed on. In the absence of a universal antigenic indication (analogous to CD34+ for HSC) and an universal assay (analogous to the repopulation assays for HSC) MSC are often identified simply by testing a cultures’ differentiation potential into colony forming units (CFU) indicative of proliferative capacity (see below) and into several lineages of mesenchymal tissue as defined above (Pittenger et al., 1999). Also, the ability to adhere to tissue culture plastic and a fibroblast-like morphology are taken as characteristic markers for MSC (Prockop, 1997).

Recently, different surface markers have been associated with MSC including D7fib (Jones et al., 2002), Stro1 (Stenderup et al., 2001), CD45 and glycophorin A (Jones et al., 2004, Pittenger et al., 1999, Reyes et al., 2001), BMPR1a (Zvaifler et al., 2000) (for comparative analysis, see Young and Black, 2004).

Further complications for comparative review arise when different sources, extraction and cultivation methods are used. Even when narrowing sources to bone marrow (as in this review), the site of extraction is reported to influence cell behaviour: e.g. MSC from alveolar bone show less chondrogenic and adipogenic potential compared to iliac bone (Matsubara et al., 2004). Isolation is usually conducted by density centrifugation (sometimes enhanced by gradient solutions) to obtain the mononuclear fraction of marrow cells and by using the widely reported ability of MSC to adhere to tissue culture plastic (Sekiya et al., 2002). Newer methods employ magnetic beads (Stenderup et al., 2001) or FACS sorting (Fickert et al., 2003) in conjunction with antibodies to the proposed MSC markers above. Additionally, widely differing standards regarding serum composition, culture conditions, and growth factor application in MSC cultivation exist. Differing conditions can lead to enrichment of different subsets of MSC with differing clonogenic potential. All these potential deviation points in current methods are summarised in Table 1.

For the purposes of this review, MSC will be defined as post-embryonic, bone-marrow derived cells, naturally capable of multipotent differentiation into connective tissue of non-haematopoietic lineage; in particular bone, ligaments, tendons, fibres, cartilage, and adipose tissue. (Compare the cell type called PPIMSC by Young and Black (2004).)

If the definition of MSC is elusive, a definition of aging is even more daunting. The patchwork nature of different research foci, coupled with fundamental uncertainties about the nature and the evolutionary role of the aging process preclude a common definition. In cytological study, further challenges arise in distinguishing between aging in vivo and prolonged cultivation in vitro that might or might not simulate ‘true’ aging (‘in vitro aging’).

We adopt a definition of aging as “the sum of primary restrictions in regenerative mechanisms of multicellular organisms” (Sames and Stolzing, 2005). This definition highlights the involvement of MSC in cell replenishment and thus in influencing lifespan.

Aging can be conceptually distinguished from senescence, with the latter emphasising the cellular level. Here, we adopt the definition of Campisi (2000) that equates senescence with replicative senescence (Hayflick and Moorhead, 1961) by defining it as “an essentially irreversible arrest of cell division”. Unlike apoptotic cells, senescent cells remain alive, despite a derangement of function (Itahana et al., 2001). Cellular senescence is a complex phenotype that entails changes in both function and replicative capacity. Different experimental protocols, culture conditions, and cell types yield different kinds of senescence. Generally, senescent cells display a characteristic enlarged, flattened morphology and are characterized by an irreversible G1 growth arrest involving the repression of genes that drive cell cycle progression and the upregulation of cell cycle inhibitors like p53/p21 and p16/RB. It was suggested that there are notable distinctions between senescent states induced by the p53 and p16/RB pathways; there is an emerging consensus that senescence occurs via one pathway or the other, with p53 mediating senescence due primarily to telomere dysfunction and DNA damage and the p16/RB pathway mediating senescence due primarily to oncogenes, chromatin disruption, and various stresses (Campisi, 2005).

It has been speculated that senescence may lead to arrested regeneration in tissues and thus to organ failure and death (Knapowski et al., 2002). One degenerative factor in senescence is the accumulation of damage in the cell (Gao et al., 2001, von Zglinicki et al., 2001). Furthermore, senescent cells secrete factors including degradative enzymes, inflammatory cytokines, and growth factors that stimulate tissue aging and tumorigenesis (Krtolica and Campisi, 2003).

Nonetheless, there are persistent doubts about the phenomenon's relevance for in vivo aging (Hornsby, 2002). Critical tissues such as cardiac or neural tissue divides little if at all. Furthermore, senescence in metazoans composed entirely of postmitotic cells is just as predictable and robust as that in metazoans containing mitotic cells (Effros et al., 2005). The immediate relevance of senescence is likely closer to cancer than to other aging-related developments.

The capacity of MSC to effect tissue and organ regeneration is still not well understood. MSC will populate a wide variety of tissues after systemic infusion (Gao et al., 2001).

Recent evidence suggests that the process of tissue repair is driven by tissue-specific progenitor cells which are replenished by MSC from bone marrow by migration–differentiation (Shake et al., 2002), fusion (Kotton et al., 2001, Prockop et al., 2003b) or the provision of a stromal support network (Alexanian and Kurpad, 2005, Sheng et al., 1998). Involvement of bone marrow-derived stem cells has been demonstrated in the regeneration of a number of organs/tissues including bone (Shirley et al., 2005), skin (Mori et al., 2005), liver (Grompe, 2005), kidney (Okada, 2005) and muscle (Natsu et al., 2004). Consequently, any loss in numbers or functionality with age would have profound consequences for the maintenance of tissue viability (Pelicci, 2004)

Section snippets

Morphology

In vitro aged MSC are reportedly bigger (Baxter et al., 2004, Mauney et al., 2004, Stenderup et al., 2003) than their young counterparts; they exhibit more podia and spread further (Mauney et al., 2004) and contain more actin stress fibers (Stenderup et al., 2003). Increase in cell size is often associated with senescence (Dimri et al., 1995, Hayflick and Moorhead, 1961).

MSC from older patients show no spindle-formed (young) MSC morphology in culture, whereas MSC from young donors exhibit the

Extrinsic and intrinsic MSC aging

When considering the link between aging and MSC, we are faced with two interrelated components: The effect of aging on MSC themselves, and the contribution of MSC to the aging of the organism. In this, a primary concern is to determine whether MSC are aging internally (intrinsic theory) or if the cells are driven into proliferative silence by changes within the surrounding tissue (extrinsic theory).

For practical reasons, aging of MSC tends to be studied in monolayer cultures of purified or

Collating findings

In collating current findings on age markers in MSC, the emerging picture is curiously consistent in its inhomogeneity. A few methodological difficulties in MSC analysis have been discussed, and in the context of aging-related research further difficulties are presented by the inclusion of differently defined age groups, variations in sex and disease status of donors.

However, evidence suggests that many of the apparent inconsistencies are likely attributable to observations made in Section 1.

References (196)

  • M.R. Dressler et al.

    Effects of age on the repair ability of mesenchymal stem cells in rabbit tendon

    J. Orthop. Res.

    (2005)
  • D. Egrise et al.

    The number of fibroblastic colonies formed from bone marrow is decreased and the in vitro proliferation rate of trabecular bone cells increased in aged rats

    Bone

    (1992)
  • S. Fickert et al.

    Identification, quantification and isolation of mesenchymal progenitor cells from osteoarthritic synovium by fluorescence automated cell sorting

    Osteoarthr. Cartilage

    (2003)
  • N.R. Forsyth et al.

    Telomerase and differentiation in multicellular organisms: turn it off, turn it on, and turn it off again

    Differentiation

    (2002)
  • A.J. Friedenstein et al.

    Bone marrow stromal colony formation requires stimulation by haemopoietic cells

    Bone Miner.

    (1992)
  • I. Garkavtsev et al.

    Molecular aspects of the relationship between cancer and aging: tumor suppressor activity during cellular senescence

    Exp. Gerontol.

    (1998)
  • A. Globerson

    Thymocytopoiesis in aging: the bone marrow-thymus axis

    Arch. Gerontol. Geriatr.

    (1997)
  • M.J. Greenwood et al.

    Telomeres, telomerase, and hematopoietic stem cell biology

    Arch. Med. Res.

    (2003)
  • C.W. Greider et al.

    Identification of a specific telomere terminal transferase activity in Tetrahymena extracts

    Cell

    (1985)
  • K.H. Han et al.

    Alteration of the TGF-beta/SMAD pathway in intrinsically and UV-induced skin aging

    Mech. Ageing Dev.

    (2005)
  • T.B. Harris et al.

    Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly

    Am. J. Med.

    (1999)
  • L. Hayflick et al.

    The serial cultivation of human diploid cell strains

    Exp. Cell Res.

    (1961)
  • A.A. Hegewald et al.

    Hyaluronic acid and autologous synovial fluid induce chondrogenic differentiation of equine mesenchymal stem cells: a preliminary study

    Tissue Cell

    (2004)
  • B. Johnstone et al.

    In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells

    Exp. Cell Res.

    (1998)
  • M. Kobune et al.

    Telomerized human multipotent mesenchymal cells can differentiate into hematopoietic and cobblestone area-supporting cells

    Exp. Hematol.

    (2003)
  • O.K. Lee et al.

    Isolation of multipotent mesenchymal stem cells from umbilical cord blood

    Blood

    (2004)
  • H.V. Leskela et al.

    Osteoblast recruitment from stem cells does not decrease by age at late adulthood

    Biochem. Biophys. Res. Commun.

    (2003)
  • L. Liu et al.

    Telomerase deficiency impairs differentiation of mesenchymal stem cells

    Exp. Cell Res.

    (2004)
  • D.B. Lombard et al.

    DNA repair, genome stability, and aging

    Cell

    (2005)
  • J.A. Martin et al.

    Roles of articular cartilage aging and chondrocyte senescence in the pathogenesis of osteoarthritis

    Iowa Orthop. J.

    (2001)
  • M.E. Martinez et al.

    Influence of skeletal site of origin and donor age on 1,25(OH)2D3-induced response of various osteoblastic markers in human osteoblastic cells

    Bone

    (1999)
  • T. Matsubara et al.

    A new technique to expand human mesenchymal stem cells using basement membrane extracellular matrix

    Biochem. Biophys. Res. Commun.

    (2004)
  • J.R. Mauney et al.

    Matrix-mediated retention of osteogenic differentiation potential by human adult bone marrow stromal cells during ex vivo expansion

    Biomaterials

    (2004)
  • J.R. Mauney et al.

    Matrix-mediated retention of adipogenic differentiation potential by human adult bone marrow-derived mesenchymal stem cells during ex vivo expansion

    Biomaterials

    (2005)
  • R.C. Meagher et al.

    An analysis of the multilineage production of human hematopoietic progenitors in long-term bone marrow culture: evidence that reactive oxygen intermediates derived from mature phagocytic cells have a role in limiting progenitor cell self-renewal

    Blood

    (1988)
  • T. Alliston et al.

    TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation

    Embo J.

    (2001)
  • R.C. Allsopp et al.

    Replicative senescence of hematopoietic stem cells during serial transplantation: does telomere shortening play a role?

    Oncogene

    (2002)
  • K.E. Armour et al.

    Defective bone formation and anabolic response to exogenous estrogen in mice with targeted disruption of endothelial nitric oxide synthase

    Endocrinology

    (2001)
  • D. Baksh et al.

    Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy

    J. Cell. Mol. Med.

    (2004)
  • M.A. Baxter et al.

    Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion

    Stem Cells

    (2004)
  • R.J. Bergman et al.

    Age-related changes in osteogenic stem cells in mice

    J. Bone Miner. Res.

    (1996)
  • G.L. Bianchi Scarra et al.

    Fibroblastoid colony-forming cells in myeloproliferative disorders

    Acta Haematol.

    (1983)
  • K.G. Brockbank et al.

    Quantitation of stromal and hemopoietic progenitors in spleen and femoral marrow derived from Steel (Slj/+ and Sl/Sld) mice and their normal littermates

    Exp. Hematol.

    (1983)
  • 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

    J. Cell Biochem.

    (1997)
  • J. Campisi

    Aging and cancer: the double-edged sword of replicative senescence

    J. Am. Geriatr. Soc.

    (1997)
  • J. Campisi

    Cancer, aging and cellular senescence

    In Vivo

    (2000)
  • E. Canalis et al.

    Bone morphogenetic proteins, their antagonists, and the skeleton

    Endocr. Rev.

    (2003)
  • D. Cheleuitte et al.

    In vitro secretion of cytokines by human bone marrow: effects of age and estrogen status

    J. Clin. Endocrinol. Metab.

    (1998)
  • C.S. Chen

    Phorbol ester induces elevated oxidative activity and alkalization in a subset of lysosomes

    BMC Cell. Biol.

    (2002)
  • C.P. Chiu et al.

    Differential expression of telomerase activity in hematopoietic progenitors from adult human bone marrow

    Stem Cells

    (1996)
  • Cited by (520)

    • Roles and mechanisms of optineurin in bone metabolism

      2024, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    View full text