You are on page 1of 10

Home Search Collections Journals About Contact us My IOPscience

Mesenchymal stem cells for tissue engineering and regenerative medicine

This article has been downloaded from IOPscience. Please scroll down to see the full text article.

2006 Biomed. Mater. 1 63

(http://iopscience.iop.org/1748-605X/1/2/003)

View the table of contents for this issue, or go to the journal homepage for more

Download details:
IP Address: 138.87.11.21
The article was downloaded on 09/12/2012 at 21:15

Please note that terms and conditions apply.


INSTITUTE OF PHYSICS PUBLISHING BIOMEDICAL MATERIALS
Biomed. Mater. 1 (2006) 63–71 doi:10.1088/1748-6041/1/2/003

Mesenchymal stem cells for tissue


engineering and regenerative medicine
Suk-Kee Tae, Seok-Hyn Lee, Jae-Sik Park and Gun-Il Im
Department of Orthopaedics, Dongguk University International Hospital, Goyang, Korea
E-mail: gunil@duih.org

Received 30 March 2006


Accepted for publication 6 April 2006
Published 26 April 2006
Online at stacks.iop.org/BMM/1/63

Abstract
Mesenchymal stem cells (MSCs) exist in bone marrow and other musculoskeletal tissues.
These cells contribute to the homeostasis of musculoskeletal tissue as well as support for the
growth and differentiation of primitive hemopoietic cells. Recent advancements in tissue
engineering and regenerative medicine have highlighted MSCs as a potential source of cells
which would differentiate to a variety of tissue tailored to individual needs. This paper briefly
outlines the current status of MSCs, focusing on their biological characteristics and potential
for clinical applications.

1. Introduction cartilage, fat and muscle [4, 5]. In addition, to meet the
high demand for precursors during tissue growth and repair,
Bone marrow contains mesenchymal stem cells (MSCs) which uncommitted progenitors can be recruited from other tissue
support hematopoietic stem cells and provide progenitors sources [6].
for several mesenchymal tissues, including bone, cartilage, The multipotential of MSCs, their easy isolation and
tendon, fat and muscle [1]. Comparable adult stem cells culture, and their high ex vivo expansive potential make
have also been isolated from a wide variety of tissues. These these cells an attractive therapeutic tool for potential use in
cells can differentiate into specialized cells with a phenotype regenerative medicine and tissue engineering.
distinct from that of the precursor under the influence of
appropriate signals. 2. In vitro characteristics of mesenchymal stem cells
MSCs were first identified in the pioneering studies of
Friedenstein, who isolated bone-forming progenitor cells from Even though MSCs can be easily isolated and expanded in
rat marrow [2]. MSCs represent a very small fraction, 0.001– culture through many generations while retaining the capacity
0.01%, of the total population of nucleated cells in marrow to differentiate, little information is currently available about
[3]. However, they can be isolated and expanded with high the biology of stem cells in vivo and their precise role in tissue
efficiency, and induced to differentiate to multiple lineages repair or regeneration. This may be due in part to the lack of
under defined culture conditions. Along with their extensive useful cell-specific markers. MSCs can differentiate in vitro
capacity for self-renewal, stem cells display a broad potential into cells of connective tissue lineages, including bone, fat,
for generating diverse differentiated progenies. and cartilage under appropriate culture conditions.
Adult stem cells had been considered to be
developmentally committed so that a kind of stem cell 2.1. Isolation and characterization of MSCs
generates specific cell lineages. This concept has been
radically challenged by several uncanny and unanticipated MSCs are generally isolated from bone marrow aspirates
discoveries. Recent studies have demonstrated that particular harvested from the superior iliac crest of the pelvis in humans
stem cells, besides producing an expected set of cell [3, 7] although other sources such as the tibial and femoral
characteristics of the tissue they reside in, also give rise marrow compartments [8, 9] and the thoracic and lumbar
to a set of totally different cells. MSCs can give rise to spine are also available [10]. Although they represent a
nonmesenchymal cells such as neural cells or epithelial cells minor fraction of the total nucleated cell population in marrow,
as well as a variety of mesenchymal phenotypes such as bone, MSCs can be plated and enriched using standard cell culture

1748-6041/06/020063+09$30.00 © 2006 IOP Publishing Ltd Printed in the UK 63


S-K Tae et al

Figure 2. Human mesenchymal stem cells from bone marrow are


strongly positive for STRO-1 antibody (left) and negative for CD-34
(right).
Figure 1. Human mesenchymal stem cells from bone marrow
growing in vitro on the floor of a plastic dish.
with non-hematopoietic progenitor bone marrow stromal cells
(figure 2) [22]. The SB-10 antibody was shown to be reactive
techniques. The marrow sample is fractionated on a density with an antigen present on undifferentiated MSCs, which
gradient solution. Then the cells are plated at densities ranging disappeared after the cells began the osteogenic differentiation
from 1 × 104 cells cm−2 to 0.4 × 106 cells cm−2 [3, 11, and express cell surface alkaline phosphatase [23]. The
12]. Cells are generally cultured in a basal medium such as specific SB-10 antigen was later identified as CD166 (activated
Dulbecco’s modified Eagle’s medium (high glucose) in the leukocyte-cell adhesion molecule, ALCAM) [23], which may
presence of 10% fetal bovine serum (FBS) [3]. MSCs have play a role in the progression of osteogenic differentiation.
a fibroblastic morphology in a monolayer culture and adhere The SH-2 antibody reacts with an epitope present on the
to the tissue culture substrate. Primary cultures are usually transforming growth factor-beta (TGF-β) receptor endoglin
maintained for 12–16 days, during which time the nonadherent (CD105) [21]. Both the SH-3 and SH-4 antibodies [24]
hematopoietic cells are removed with the exchange of the apparently recognize distinct epitopes on the membrane-bound
medium. The addition of fibroblast growth factor-2 (FGF-2) ecto-5-nucleotidase (CD73) [21]. SH-2, 3, 4 antibodies do
to primary cultures of human MSCs leads to an enhanced not react with hematopoietic cells or with osteocytes [24].
proliferation and osteogenic potential [13]. Although plastic Regrettably, all of the antigens are also expressed on a host
adherence remains the principal method to isolate MSCs, of other cell types and do not provide the specificity needed
immunodepletion methods involving CD34/CD45/CD11b to extend these studies to in vivo evaluation of tissue-specific
may be used to generate purified MSC preparations [14]. stem cells.
By light or phase contrast microscopy, MSC cultures MSCs express a large spectrum of cell adhesion molecules
display a rather homogenous population of fibroblast-like cells that have potential importance in cell binding and homing
(figure 1). While a small fraction of MSCs actively proliferate interactions. CD44, a receptor for various ligands such as
(approximately 10% at S + G2 + M), the vast majority of hyaluronan and osteopontin, is also expressed and particularly
cells are standing at the G0/G1 phase of the cell cycle [15]. [15] plays a central role in the organization of the extracellular
After multiple passages, MSCs show a large but exceedingly matrix in the marrow or in the bone [25, 26]. Expression of
erratic expansive potential. MSCs cannot be expanded over specific integrins also plays a role in homing to sites of injury
30–40 doublings [16, 17]. This is determined by several and binding to specific matrix molecules [27].
factors: the procedures for harvesting the marrow [16–18];
the frequency of MPC in marrow harvests (2–5 MPC per
1 × 106 mononuclear cells) [19]; the age or condition of the 2.3. Differentiation of MSCs
donor [20]. After extensive subcultivation impairs, cells show
Osteogenic activation requires β-glycerol-phosphate, ascorbic
evident signs of senescence and apoptosis [15].
acid-2-phosphate, dexamethasone and fetal bovine serum.
When cultured in a monolayer in the presence of these
2.2. Surface markers of MSCs
supplements, the cells acquire an osteoblastic morphology
In order to characterize MSCs, the antigenic phenotypes with upregulation of the alkaline phosphatase activity and
of MSCs have been studied. The antigenic types are not deposition of a calcium-rich mineralized extracellular matrix.
unique, and also vary with subcultures. It shows features Chondrogenic differentiation occurs when MSCs are
of mesenchymal, endothelial, epithelial and muscle cells. grown in a three-dimensional culture format, a serum-free
In general, MSCs do not express the typical hematopoietic nutrient medium and the addition of a member of the TGF-
antigens such as CD45, CD34 and CD14 [15, 21]. There are β super-family such as TGF-β1, β2, β3 or BMPs. When
several antibodies that were extensively investigated by several these conditions are met the cells initiate an expression of a
groups. STRO-1 was identified as an antibody that reacted number of cartilage-specific extracellular matrix components

64
Mesenchymal stem cells for tissue engineering and regenerative medicine

and a spectacular change in cell morphology. TGF- phenotypes with characteristics of adipocytes, osteoblasts,
β1, β2 and β3 have the ability to induce this response chondrocytes, smooth muscle cells, skeletal myotubes and
although TGF-β2 and 3 are more effective than β1 in cardiomyocytes when treated with dexamethasone [51]. It
promoting chondrogenesis [28]. During differentiation in has not been confirmed whether muscle- and bone marrow-
the presence of TGF-β, MSCs synthesize all components of resident mesenchymal stem cells represent the same kind of
the normal articular cartilage matrix (aggrecan, link protein, progenitor. However, the work by Ferrari et al [6] has provided
fibromodulin, cartilage oligomeric matrix protein, decorin, strong evidence that the populations of muscle progenitors
type II collagen) [28]. present in skeletal muscle are derived from uncommitted
MSCs cultured in a monolayer in the presence of bone marrow mesenchymal progenitors and are different from
isobutylmethylxanthine become adipocytes with large lipid- muscle satellite cells.
filled vacuoles. Adipogenic differentiation of MSCs is induced
by the nuclear receptor and transcription factor, peroxisome 3.2. Bone-derived MSC
proliferator-activated receptor-gamma (PPAR-γ ). Both IL-1
and TNF-α suppress adipogenesis, which is mediated through Four cellular subsets were sorted from primary cultures of
NF-κB activated by the TAK1/TAB1/NF-κB induction kinase human bone, depending on expression of the stromal precursor
cascade [29]. The effect of inhibition by these cytokines cell marker STRO-1 and the osteoblastic marker alkaline
leads to differentiation toward osteogenesis. Induction of phosphatase (ALP) [52]. The STRO-1+/ALP-subset showed
myogenesis by 5-azacytidine was reported for embryonic and a preosteoblastic phenotype with a reduced ability to form a
adult cells [30, 31]. mineralized bone matrix and the lack of expression of bone
Treatment of MSCs with isobutylmethylxanthine and sialoprotein, osteopontin and parathyroid hormone receptor.
dibutyryl cyclic AMP induced expression of early markers of Only these subpopulation cells gave rise to all of the four
neuronal differentiation [32]. Transdifferentiation of mouse subsets of STRO-1/ALP cells present in the primary culture.
marrow stromal-derived mature osteoblasts and the stromal Thus, these results have demonstrated that cultures of human
cells themselves to a neural phenotype was achieved by bone yield osteoprogenitors as well as end-stage differentiated
treatment with 5-azacytidine in the presence of nerve growth osteoblasts.
factor, BDNF and neurotrophin-3 [33]. Treatment of rat Studies with nonimmortalized clonal cell lines derived
cells with DMSO/butylated hydroxyanisole in the presence from human trabecular bone have shown that bipotent-
of bFGF and PDGF was also successful in inducing a neural (osteo/adipo) committed progenitors are also present. Thus,
phenotype [34]. uncommitted mesenchymal stem cells are also ubiquitously
positioned in bones which, under appropriate stimuli, may self-
3. Tissue-specific mesenchymal stem cells renew, commit and generate cells exhibiting the phenotypic
and functional characteristics of the bone or adipose
In addition to marrow, other sources of stem cells with tissue [53].
mesenchymal potential include periosteum [35, 36], trabecular
bone [37, 38], adipose tissue [39–41], synovium [42], skeletal 3.3. Adipose tissue-derived MSC
muscle [31], lung [43] and deciduous teeth [44].
Adipose tissue stromal cells contain adipocyte progenitors
at various stages of maturity, including the stromal-vascular
3.1. Muscle-derived MSC
(SV) cells, considered as the less differentiated tissue-
Work performed with adult human skeletal muscle has resident adipocyte progenitor. Those cells proliferate and
demonstrated the existence of cells with properties of early differentiate into mature adipocytes during cold acclimation
myogenic progenitors [45]. A mixture of stellate-shaped [54] and after caloric excess [55]. SV cells are a class of
cells and multinucleated myotubes was formed after tissue fat-resident-committed mesenchymal progenitors, exhibiting
enzymatic dissociation and cell cultivation. Cells showed at least a bipotent differentiation potential, since they can
signs of differentiation when dexamethasone was added differentiate into adipocytes or chondrocytes [56]. They can
[46]. The differentiated population included cells with the be induced to differentiate into adipocytes by glucocorticoids
phenotype of skeletal and smooth muscle, bone, cartilage and [57]. Stem cells from adipose tissue, variously referred
fat. In addition, there was a minor cellular subset that contains to as processed lipoaspirate (PLA) cells [40], have been
slowly dividing cells in culture but rapidly after grafting shown to have similar differentiation potentials [39]. There
was preset. The subset probably represents uncommitted is a little difference between cells from marrow and fat in
mesenchymal stem cells that persist in the environment of terms of yield, growth kinetics, cell senescence, multi-lineage
the recipient muscle. These cells [47–49] seem to be different differentiation capacity and gene transduction efficiency. The
from muscle satellite cells that are considered as the muscle utility of these cells in therapeutic applications may then
stem cell [50]. depend on the availability of tissue specimens and the ease of
Cells from myocardium also exhibit properties of in vitro expansion. However, recent works on the osteogenic
mesenchymal progenitors. It has been reported that and chondrogenic potentials of adipose-derived MSCs show
cultures of neonatal rat heart gives rise to a population inferior capacity for osteogenesis or chondrogenesis compared
of adherent cells which generate several mesenchymal with bone marrow-derived MSCs [58, 59].

65
S-K Tae et al

4. Mobilization of mesenchymal stem cells MSCs have also been shown to engraft at high levels
in lung tissue following exposure to bleomycin offering
Bone marrow stroma can feed progenitors into distant tissues protection against bleomycin-induced lung injury, including
[1]. Thus, the mesenchymal stem cell must leave the marrow inflammation and collagen deposition.
stroma for destinations in other tissues. Cell-to-cell or cell- These observations have broad implications in the area of
to-matrix interactions between mesenchymal progenitors and lung disease associated with environmental damage [14].
stromal components should loosen up in order to facilitate Stem cells with the ability to differentiate into neurons,
the entrance of the progenitor into the blood stream. Then astrocytes and oligodendrocytes have been isolated from rat
peripheral blood should provide a trail route for mesenchymal spinal cord [72], and implantation of neural stem cells in an
stem cells in the search of their final destinations where they adult rat model of spinal cord injury resulted in long-term
are supposed to home, expand and further differentiate. functional improvement [71]. The ability of bone marrow-
In addition to adult bone marrow and other mesenchymal derived MSCs to differentiate into neural lineages in vitro
tissues, umbilical cord blood is also a source of mesenchymal and after transplantation in both mice and rats provoked the
progenitors [60]. These ‘in motion’ mesenchymal cells expectation that they may be useful in the treatment of stroke,
display many common features with adult human bone traumatic injury and Parkinson’s disease. Furthermore, it was
marrow-derived mesenchymal progenitors, such as adhesion recently verified that adult human bone marrow cells could
to plastic, morphology, expression of cell membrane and enter the brain and generate neurons after transplantation [73].
cytoplasmic antigens, and a potential to differentiate into In orthopedics, there are many applications involving
osteo/chondrogenic and adipogenic phenotypes. Moreover,
local delivery of MSCs, which include repair of focal defects
the identification in cultures of mesenchymal cells from cord
in articular cartilage [74, 75], tendon [76], ligament [77],
blood of a subset (5–10%) of quiescent cells suggests that
intervertebral disk [78], spine fusion [79], the repair of
an uncommitted mesenchymal progenitor circulates during
segmental bone defects [80] and craniotomy defects [81]. In
gestation. The inverse correlation between content of
an animal model of osteoarthritis, delivery of stem cells by
mesenchymal progenitors in cord blood and gestational age,
intraarticular injection resulted in engraftment of those cells
a trend also observed for hematopoietic progenitors [61],
on the meniscus, fat pad and synovium with regeneration of
suggests that mesenchymal cells travel from fetal sites into
meniscal tissue and protection of the cartilage.
other tissues early during development [60].

5. Therapeutic applications of mesenchymal stem 5.2. Systemic infusion


cells in regenerative medicine A selective homing follows the infusion of mesenchymal
progenitors from bone marrow into marrow stromal sites [82].
As adult stem cells have presented several promising features
The homed MSCs facilitate engraftment and differentiation
for the development of new cell therapies, researchers have
of hematopoietic stem cells, which improves the function of
pursued a broad range of investigations for their therapeutic
hematopoietic-supporting stroma [83, 84].
utilization. MSCs can be locally applied for regeneration or
Although the issue of transplantation has been addressed
infused systematically depending on the purpose.
in several studies, results are rather contradictory in
establishing the origin (host or donor) of mesenchymal
5.1. Direct loading progenitors after allogeneic transplantation of marrow harvests
Direct loading is best suited to a clinical strategy oriented to [19, 20, 85–88]. In addition, it has been demonstrated that
augment local repair or regeneration of bone [62–64], cartilage stemness of the grafted cells determines repopulation of the
[65], tendon [66] and fat [67, 68]. Moreover, therapeutic damaged tissue [89, 90]. It is not certain whether the same
use of marrow-derived MSCs addresses a broad spectrum of occurs after transplantation of ex vivo expanded mesenchymal
indications, including cardiovascular repair, treatment of lung progenitors since progenitor stemness and function diverge as
fibrosis and spinal cord injury. cells are subcultivated [15].
Locally delivered bone marrow cells can generate de novo Systemic infusion of ex vivo expanded autologous
myocardium, indicating that stem cell therapy can be useful in mesenchymal progenitors is feasible and safe in the short
treating coronary artery disease [69]. The practical utility of term [82, 91]. On the other hand, the low immunogenic
this approach was tested in a study involving the delivery of nature of MSCs allows broad application in terms of allogeneic
bone marrow cells into the infarct zone in patients following therapy. There are several reports describing the clinical use
myocardial infarction [70]. A dramatic improvement in global of allogeneic donor-mismatched cells with little evidence of
heart function was achieved as a result of this treatment. host immune rejection or GVHD. Allogeneic bone marrow
Another study showed that engraftment of bone marrow- transplantation in children with osteogenesis imperfecta
derived cardiomyocytes in the adult heart following bone results in impressive histological changes in trabecular bone,
marrow transplantation demonstrated that MSCs are tolerated which are indicative of new dense bone formation [92].
in a xenogeneic environment while retaining their ability Engraftment of allogeneic MSCs has also been demonstrated
to be recruited to the injured myocardium and undergo in a patient with severe idiopathic aplastic anemia with
differentiation to a cardiac phenotype [71]. improvement of marrow stromal function [93].

66
Mesenchymal stem cells for tissue engineering and regenerative medicine

6. Application of mesenchymal stem cells for of large quantity of cells for transplantation and ethical and
cartilage tissue engineering legal concerns. For example, committed chondrocytes isolated
from hyaline cartilage by enzymatic digestion have been
Damage to cartilage is of great clinical consequence because tested extensively as a cell source to be injected into the
it has a poor regenerative capacity and its replacement tissue, damaged articular cartilage site for repair. However, their wide
fibrocartilage, is mechanically inferior to normal hyaline. usage in clinical trials and tissue engineering applications has
Due to the lack of blood supply, the subsequent wound been held up because of the loss of redifferentiation capability
healing response and the lack of the intrinsic ability of native after relatively limited in vitro expansion. In contrast, ES cells
chondrocytes to participate in regeneration at the injury site derived from the inner cell mass of the embryonic blastocyst
damage to cartilage results in incomplete repair. have infinite proliferation potential and are able to generate
The current treatment options (e.g., surgical intervention) cell characteristics of all three germ layers under appropriate
to repair damaged articular cartilage are less than satisfactory, inductive conditions. However, the ethical and legal concerns
and rarely restore full function or return the damaged tissue to associated with ES cells and their tumorigenic potential make
its normal biologic state. them a less ideal cell source in basic research and clinical
The final treatment for articular cartilage reconstruction applications. On the other hand, adult stem cells (e.g., MSC)
is joint replacement arthroplasty [94, 95]. Although total derived from various tissues (e.g., BM, fat and trabecular
joint replacement has advanced during the last 40 years, it bone) demonstrate a promising future in regenerative medicine
is still fraught with a number of drawbacks such as infection, because of their ability of self-renewal and differentiation
loosening and periprosthetic osteolysis. along specific lineages upon stimulation.
On the other hand, the low degree of vascularization
in cartilage makes it an ideal target for tissue engineering.
6.2. Scaffolds for cartilage tissue engineering
Tissue engineering combines the principles of engineering
and biology to design and fabricate constructs to expedite the In addition to selecting an appropriate source of cells for
repair and regeneration of damaged tissue. The underlying cartilage tissue engineering, the design of an engineered
principle of tissue engineering involves the utilization of cartilage construct, i.e. the scaffolding material in which
biocompatible and mechanically suitable scaffolds, combined the cells will reside, needs careful consideration. The
with an appropriate source of cells and bioactive molecules to physical properties (e.g., architecture, macro/microporosity,
promote the differentiation and maturation of the cell type interconnecting porosity and topography) of an engineered
of interest. These components, when combined, form a tissue substitute will play an integral role in altering its
tissue-engineered construct, which can function as the tissue biological performance. Therefore, an ideal tissue engineering
replacement material and, in principle, facilitate a faster rate scaffold should possess a number of key properties, including
of tissue repair. Tissue engineering may offer great promise being biocompatible, biodegradable, porous, mechanically
for the regeneration of cartilage. stable, cell permissive and conducive to extracellular
One of the great challenges facing cartilage tissue matrix production and deposition and biomolecular signal
engineering is designing constructs that mimic the unique transmission. Natural materials used to produce a bioactive
multiphasic cellular architecture and mechanical properties of scaffold for stem-cell-derived cartilage include agarose,
native cartilage tissue. As such, successful cell-based cartilage alginate, hyaluronic acid, gelatin, fibrin glue and collagen
tissue engineering will depend on not only the selection of derivatives. The principal disadvantage of these constructs is
an appropriate biocompatible scaffolding material, but also their mechanical fragility, which severely restricts their clinical
materials with suitable mechanical properties that will result usefulness. The mechanical and biochemical qualities and the
in a desirable clinical outcome when combined with cells and degradation of synthetic biomaterials are more easily modified
bioactive molecules. than those of natural polymers. They can be fabricated as
needed and can be manufactured with more uniform properties
6.1. Cells for cartilage tissue engineering and purity. Polyglycolic acid is a long-standing, well-studied
Selecting an ideal source of cells for cartilage tissue scaffold for engineering neocartilage, but degrades at a faster
engineering requires the achievement of a number of criteria, rate and is weaker than most synthetic scaffolds [96, 97]. The
which include easy access and availability of the source cells, polylactides are slow absorbing polymers. Polylactic acid
an extensive self-renewal or expansion capability of the source press-coated with human MSCs has been shown to support
cells, a capacity to differentiate readily into cell lineages of chondrogenesis in vitro [98, 99]. Poly(lactic-co-glycolic acid)
interest upon instructive signal, and a lack of or minimal and poly(ethylene glycol) blended into a polymeric foam of
immunogenic or tumorigenic ability of the source cells. ratio 80/20, polydioxanone, and the copolymer poly(L-lactide-
Over the years, three major cell types have been examined E-caprolactone) prepared as a nanofibrous scaffold are also
for their potential application in cartilage repair and tissue reported to be useful scaffolds for the chondrogenic induction
engineering, namely committed chondrocytes, embryonic of human MSCs (figure 3) [100].
stem (ES) cells and adult stem cells. Each of these cell Hydrogels are developed with a view to being customized
types has demonstrated its limitations and advantages over for small, complex geometrical defects typically exhibited
the others due to their intrinsic biological properties, such as in facial structures. These can fill tissue defects using
their proliferative and differentiation potentials, availability minimally invasive procedures and be molded to the specific

67
S-K Tae et al

[7] Digirolamo C M, Stokes D, Colter D, Phinney D G, Class R


and Prockop D J 1999 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 Br. J. Hematol.
107 275–81
[8] Murphy J M, Dixon K, Beck S, Fabian D, Feldman A and
Barry F 2002 Reduced chondrogenic and adipogenic
activity of mesenchymal stem cells from patients with
advanced osteoarthritis Arthritis Rheum. 46 704–13
[9] Oreffo R O, Bord S and Triffitt J T 1998 Skeletal progenitor
cells and ageing human populations Clin. Sci. 94 549–55
[10] D’Ippolito G, Schiller P C, Ricordi C, Roos B A and
Figure 3. Repair of an osteochondral defect with the implantation Howard G A 1999 Age-related osteogenic potential of
of the composite of MSCs and polydioxanone scaffold in the rabbit mesenchymal stromal stem cells from human vertebral bone
(left), control without implantation (right). Safranin-O staining marrow J. Bone Miner. Res. 14 1115–22
(×40). [11] Lodie T A, Blickarz C E, Devarakonda T J, He C, Dash A B,
Clarke J, Gleneck K, Shihabuddin L and Tubo R 2002
Systematic analysis of reportedly distinct populations of
shape. Photopolymerizing poly(ethylene glycol)-diacrylate
multipotent bone marrow-derived stem cells reveals a lack
is a recently investigated system for engineering osteogenic of distinction Tissue Eng. 8 739–75
and chondrogenic cells from MSCs [101–103]. A variety of [12] McBride C, Gaupp D and Phinney D G 2003 Quantifying
hybrid scaffolds using both synthetic and natural polymers can levels of transplanted murine and human mesenchymal
be used for the effective delivery of MSCs. They combine the stem cells in vivo by real-time PCR Cytotherapy 5 7–18
better biocompatibility of natural polymers with the strength [13] Martin I, Muraglia A, Campanile G, Cancedda R and Quarto R
1997 Fibroblast growth factor-2 supports ex vivo expansion
and moldable nature of synthetic polymers. Problems with and maintenance of osteogenic precursors from human
purity, isolation and supply of the natural component remain bone marrow Endocrinology 138 4456–62
a concern. [14] Ortiz L A, Gambelli F, McBride C, Gaupp D, Baddoo M,
Kaminski N and Phinney D G 2003 Mesenchymal stem cell
engraftment in lung is enhanced in response to bleomycin
7. Conclusion exposure and ameliorates its fibrotic effects Proc. Natl
Acad. Sci. 100 8407–11
Recent enthusiasm for stem cells has contributed to the [15] Conget P A and Minguell J J 1999 Phenotypical and functional
current knowledge in the biology and clinical application of properties of human bone marrow mesenchymal progenitor
MSCs. However, this information is not yet sufficient for cells J. Cell. Physiol. 181 67–73
establishing MSCs as safe and effective therapeutic tools for [16] Bruder S P, Jaiswal N and Haynesworth S E 1997 Growth
kinetics, selfrenewal, and the osteogenic potential of
tissue regeneration. Although much has been learnt about purified human mesenchymal stem cells during extensive
MSCs in the past few years, much more remains to be subcultivation and following cryopreservation J. Cell.
discovered in the future. A multidisciplinary approach would Biochem. 64 278–94
be necessary for the best effort to accomplish the goal of [17] Phinney D G, Kopen G, Righter W, Webster S, Tremain N and
using MSCs as the universal ‘stuff’ in tissue engineering and Prockop D J 1999 Donor variation in the growth properties
and osteogenic potential of human marrow stromal cells
regenerative medicine.
J. Cell. Biochem. 75 424–36
[18] Blazsek I, Delmas Marsalet B, Legras S, Marion S,
References Machover D and Misset J L 1999 Large-scale recovery and
characterization of stromal cellassociated primitive
[1] Caplan A I 1994 The mesengenic process Clin. Plast. Surg. 21 haemopoietic progenitor cells from filterretained human
429–35 bone marrow Bone Marrow Transplant. 23 647–57
[2] Friedenstein A J P S and Petrakova K V 1966 Osteogenesis in [19] Koc O N et al 1999 Bone marrow-derived mesenchymal stem
transplants of bone marrow cells J. Embryol. Explor. cells remain host-derived despite successful hematopoietic
Morph. 16 381–90 engraftment after allogeneic transplantation in patients with
[3] Pittenger M F, Mackay A M, Beck S C, Jaiswal R K, lysosomal and peroxisomal storage diseases Exp. Hematol.
Douglas R, Mosca J D, Moorman M A, Simonetti D W, 27 1675–81
Craig S and Marshak D R 1999 Multilineage potential of [20] Galotto M et al 1999 Stromal damage as consequence of
adult human mesenchymal stem cells Science 284 143–7 high-dose chemo/radiotherapy in bone marrow transplant
[4] Kopen G C, Prockop D J and Phinney D G 1999 Marrow recipients Exp. Hematol. 27 1460–6
stromal cells migrate throughout forebrain and cerebellum, [21] Pittenger M F, Mackay A M, Beck S C, Jaiswal R K,
and they differentiate into astrocytes after injection into Douglas R, Mosca J D, Moorman M A, Simonetti D W,
neonatal mouse brains Proc. Natl Acad. Sci. USA Craig S and Marshak D R 1999 Multilineage potential of
96 10711–6 adult human mesenchymal stem cells Science 284 143–7
[5] Reyes M and Verfaillie C M 1999 Turning marrow into brain: [22] Simmons P J and Torok-Storb B 1991 Identification of stromal
generation of glial and neuronal cells from adult bone cell precursors in human bone marrow by a novel
marrow mesenchymal stem cells Blood 94 377a monoclonal antibody, STRO-1 Blood 78 55–62
[6] Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, [23] Bruder S P, Kraus K H, Goldberg V M and Kadiyala S 1998
Stornaiuolo A, Cossu G and Mavilio F 1998 Muscle The effect of implants loaded with autologous
regeneration by bone marrow-derived myogenic progenitors mesenchymal stem cells on the healing of canine segmental
Science 279 1528–30 bone defects J. Bone Joint Surg. Am. 80 985–96

68
Mesenchymal stem cells for tissue engineering and regenerative medicine

[24] Haynesworth S E, Baber M A and Caplan A I 1992 Cell human infrapatellar fat pads J. Bone Joint Surg. Br. 85
surface antigens on human marrow-derived mesenchymal 740–7
cells are detected by monoclonal antibodies Bone [41] Wickham M Q, Erickson G R, Gimble J M, Vail T P and
13 69–80 Guilak F 2003 Multipotent stromal cells derived from the
[25] Minguell J J 1993 Is hyaluronic acid the ‘organizer’ of the infrapatellar fat pad of the knee Clin. Orthop. 196–212
extracellular matrix in marrow stroma? Exp. Hematol. 21 [42] De Bari C, Dell’Accio F, Tylzanowski P and Luyten F P 2001
7–8 Multipotent mesenchymal stem cells from adult human
[26] Yamazaki M, Nakajima F, Ogasawara A, Moriya H, synovial membrane Arthritis Rheum. 44 1928–42
Majeska R J and Einhorn T A 1999 Spatial and temporal [43] Noort W A et al 2002 Mesenchymal stem cells promote
distribution of CD44 and osteopontin in fracture callus engraftment of human umbilical cord blood-derived
J. Bone Joint Surg. Br. 81 508–15 CD34(+) cells in NOD/SCID mice Exp. Hematol.
Yamazaki M, Nakajima F, Ogasawara A, Moriya H, Majeska 30 870–8
R J and Einhorn T A 1999 Spatial and temporal distribution [44] Miura M, Gronthos S, Zhao M, Lu B, Fisher L W, Robey P G
of CD44 and osteopontin in fracture callus J. Bone Joint and Shi S 2003 SHED: stem cells from human exfoliated
Surg. Br. 27 1460–6 deciduous teeth Proc. Natl Acad. Sci. 100 5807–12
[27] Bogenrieder T and Herlyn M 2003 Axis of evil: molecular [45] Williams J T, Southerland S S, Souza J, Calcutt A F and
mechanisms of cancer metastasis Oncogene 22 6524–36 Cartledge R G 1999 Cells isolated from adult human
[28] Barry F, Boynton R E, Liu B and Murphy J M 2001 skeletal muscle capable of differentiating into multiple
Chondrogenic differentiation of mesenchymal stem cells mesodermal phenotypes Am. Surg. 65 22–6
from bone marrow: differentiation-dependent gene [46] Grigoriadis A E, Heersche J N and Aubin J E 1988
expression of matrix components Exp. Cell Res. Differentiation of muscle, fat, cartilage, and bone from
268 189–200 progenitor cells present in a bonederived clonal cell
[29] Suzawa M et al 2003 Cytokines suppress adipogenesis and population: effect of dexamethasone J. Cell
PPAR-gamma function through the TAK1/TAB1/NIK Biol. 106 2139–51
cascade Nature Cell Biol. 5 224–30 [47] Yoshida N, Yoshida S, Koishi K, Masuda K and Nabeshima Y
[30] Taylor S M and Jones P A 1982 Changes in phenotypic 1998 Cell heterogeneity upon myogenic differentiation:
expression in embryonic and adult cells treated with down-regulation of MyoD and Myf-5 generates’reserve
5-azacytidine J. Cell. Physiol. 111 187–94 cells J. Cell Sci. 111 769–79
[31] Wakitani S, Saito T and Caplan A I 1995 Myogenic cells [48] Baroffio A, Bochaton-Piallat M L, Gabbiani G and Bader C R
derived from rat bone marrow mesenchymal stem cells 1995 Heterogeneity in the progeny of single human muscle
exposed to 5-azacytidine Muscle Nerve 18 1417–26 satellite cells Differentiation 59 259–68
[32] Deng W, Obrocka M, Fischer I and Prockop D J 2001 In vitro [49] Gross J G and Morgan J E 1999 Muscle precursor cells
differentiation of human marrow stromal cells into early injected into irradiated mdx mouse muscle persist after
progenitors of neural cells by conditions that increase serial injury Muscle Nerve 22 174–85
intracellular cyclic AMP Biochem. Biophys. Res. Commun. [50] Schultz E 1996 Satellite cell proliferative compartments in
282 148–52 growing skeletal muscles Dev. Biol. 175 84–94
[33] Kohyama J, Abe H, Shimazaki T, Koizumi A, Nakashima K, [51] Warejcka D J, Harvey R, Taylor B J, Young H E and
Gojo S, Taga T, Okano H, Hata J and Umezawa A 2001 Lucas P A 1996 A population of cells isolated from rat
Brain from bone: efficient ‘meta-differentiation’ of marrow heart capable of differentiating into several mesodermal
stroma-derived mature osteoblasts to neurons with Noggin phenotypes J. Surg. Res. 62 233–42
or a demethylating agent Differentiation 68 235–44 [52] Gronthos S, Zannettino A C, Graves S E, Ohta S, Hay S J and
[34] Woodbury D, Reynolds K and Black I B 2002 Adult bone Simmons P J 1999 Differential cell surface expression of
marrow stromal stem cells express germline, ectodermal, the STRO-1 and alkaline phosphatase antigens on discrete
endodermal, and mesodermal genes prior to neurogenesis developmental stages in primary cultures of human bone
J. Neurosci. Res. 69 908–17 cells J. Bone. Miner. Res. 14 47–56
[35] Fukumoto T, Sperling J W, Sanyal A, Fitzsimmons J S, [53] Liu F, Malaval L, Gupta A K and Aubin J 1994 Simultaneous
Reinholz G G, Conover C A and O’Driscoll S W 2003 detection of multiple bone-related mRNAs and protein
Combined effects of insulin-like growth factor-1 and expression during osteoblast differentiation: polymerase
transforming growth factor-beta1 on periosteal chain reaction and immunocytochemical studies at the
mesenchymal cells during chondrogenesis in vitro single cell level Dev. Biol. 166 220–34
Osteoarthritis Cartilage 11 55–64 [54] Bukowiecki L J, Geloen A and Collet A J 1986 Proliferation
[36] O’Driscoll S W, Saris D B, Ito Y and Fitzimmons J S 2001 and differentiation of brown adipocytes from interstitial
The chondrogenic potential of periosteum decreases with cells during cold acclimation Am. J. Physiol. 250
age J. Orthop. Res. 19 95–103 C880–7
[37] Noth U, Osyczka A M, Tuli R, Hickok N J, Danielson K G and [55] Rajkumar K, Modric T and Murphy L J 1999 Impaired
Tuan R S 2002 Multilineage mesenchymal differentiation adipogenesis in insulin-like growth factor binding protein-1
potential of human trabecular bone-derived cells J. Orthop. transgenic mice J. Endocrinol. 162 457–65
Res. 20 1060–9 [56] Loncar D 1992 Ultrastructural analysis of differentiation of rat
[38] Tuli R, Seghatoleslami M R, Tuli S, Wang M L, Hozack W J, endoderm in vitro: adipose vascular-stromal cells induce
Manner P A, Danielson K G and Tuan R S 2003 A simple, endoderm differentiation, which in turn induces
high-yield method for obtaining multipotential differentiation of the vascular-stromal cells into
mesenchymal progenitor cells from trabecular bone chondrocytes J. Submicrosc. Cytol. Pathol. 24 509–19
Mol. Biotech. 23 37–49 [57] Park S R, Oreffo R O and Triffit J T 1999 Interconversion
[39] De Ugarte D A et al 2003 Comparison of multi-lineage cells potential of cloned human marrow adipocytes in vitro
from human adipose tissue and bone marrow Cells Tissues Bone 24 549–54
Organs 174 101–9 [58] Im G I, Shin Y W and Lee K B 2005 Do adipose
[40] Dragoo J L, Samimi B, Zhu M, Hame S L, Thomas B J, tissue-derived mesenchymal stem cells have the same
Lieberman J R, Hedrick M H and Benhaim P 2003 osteogenic and chondrogenic potential as bone
Tissue-engineered cartilage and bone using stem cells from marrow-derived cells? Osteoarthritis Cartilage 13 845–53

69
S-K Tae et al

[59] Winter A, Breit S, Parsch D, Benz K, Steck E, Hauner H, application to anterior cruciate ligament tissue engineering
Weber R M, Ewerbeck V and Richter W 2003 Cartilage-like Cell Transplant. 14 763–73
gene expression in differentiated human stem cell spheroids: [78] Richardson S M, Hughs N I, Freemont A J, Hunt J A and
a comparison of bone marrow-derived and adipose Hoyland J A Mesenchymal stem cell differentiation in a
tissue-derived stromal cells Arthritis Rheum. 48 418–29 chitosan/glycerophosphate hydrogel: implications for
[60] Erices A, Conget P and Minguell J J 2000 Mesenchymal tissue engineering of the intervertebral disc Int. J. Exp.
progenitor cells in human umbilical cord blood Br. J. Pathol. 87 A20
Haematol. 109 235–42 [79] Muschler G F, Nitto H, Matsukura Y, Boehm C, Valdevit A,
[61] Shields L E and Andrews R G 1998 Gestational age changes in Kambic H, Davros W, Powell K and Easley K 2003 Spine
circulating CD34+ hematopoietic stem/progenitor cells in fusion using cell matrix composites enriched in bone
fetal cord blood Am. J. Obstet. Gynecol. 178 931–7 marrow-derived cells Clin. Orthop. 407 102–18
[62] Goshima J, Goldberg V M and Caplan A I 1991 The origin of [80] Quarto R, Mastrogiacomo M, Cancedda R, Kutepov S M,
bone formed in composite grafts of porous calcium Mukhachev V, Lavroukov A, Kon E and Marcacci M 2001
phosphate ceramic loaded with marrow cells Clin. Orthop. Repair of large bone defects with the use of autologous
269 274–83 bone marrow stromal cells New Eng. J. Med. 344 385–6
[63] Kadiyala S, Young R G, Thiede M A and Bruder S P 1997 [81] Krebsbach P H, Mankani M H, Satomura K, Kuznetsov S A
Culture expanded canine mesenchymal stem cells possess and Robey P G 1998 Repair of craniotomy defects using
osteochondrogenic potential in vivo and in vitro Cell bone marrow stromal cells Transplantation 66 1272–8
Transplant. 6 125–34 [82] Hardy C L 1995 The homing of hematopoietic stem cells to
[64] Richards M, Huibregtse B A, Caplan A I, Goulet J A and the bone marrow Am. J. Med. Sci. 309 260–6
Goldstein S A 1999 Marrow-derived progenitor cell [83] Koc O N, Gerson S L, Cooper B W, Dyhouse S M,
injections enhance new bone formation during distraction Haynesworth S E, Caplan A I and Lazarus H M 2000 Rapid
J. Orthop. Res. 17 900–8 hematopoietic recovery after coinfusion of
[65] Johnstone B and Yoo J U 1999 Autologous mesenchymal autologous-blood stem cells and culture-expanded marrow
progenitor cells in articular cartilage repair Clin. Orthop. mesenchymal stem cells in advanced breast cancer patients
367 S156–62 receiving high-dose chemotherapy J. Clin. Oncol. 18 307–16
[66] Young R G, Butler D L, Weber W, Caplan A I, Gordon S L [84] Almeida-Porada G, Porada C D, Tran N and Zanjani E D 2000
and Fink D J 1998 Use of mesenchymal stem cells in a Cotransplantation of human stromal cell progenitors into
collagen matrix for Achilles tendon repair J. Orthop. preimmune fetal sheep results in early appearance of human
Res. 16 406–13 donor cells in circulation and boosts cell levels in bone
[67] Neubauer M et al 2005 Adipose tissue engineering based on marrow at later time points after transplantation Blood 95
mesenchymal stem cells and basic fibroblast growth factor 3620–7
in vitro Tissue Eng. 11 1840–51 [85] Keating A, Singer J W, Killen P D, Striker G E, Salo A C,
[68] Choi Y S, Park S N and Suh H 2005 Adipose tissue Sanders J, Thomas E D, Thorning D and Fialkow P J 1982
engineering using mesenchymal stem cells attached to Donor origin of the in vitro haematopoietic
injectable PLGA spheres Biomaterials 26 5855–63 microenvironment after marrow transplantation in man
[69] Orlic D et al 2001 Bone marrow cells regenerate infarcted Nature 298 280–3
myocardium Nature 410 701–5 [86] Simmonds P J, Przepiorka E D, Thomas E D and
[70] Stamm C, Westphal B, Kleine H D, Petzsch M, Kittner C, Torok-Storb B 1987 Host origin of marrow stromal cells
Klinge H, Schumichen C, Nienaber C A, Freund M and following allogeneic bone marrow transplantation
Steinhoff G 2003 Autologous bone-marrow stem-cell Nature 328 429–32
transplantation for myocardial regeneration Lancet [87] Pereira R F, O’Hara M D, Laptev A V, Halford K W,
361 45–6 Pollard M D, Class R, Simon D, Livezey K and Prockop D J
[71] Teng Y D, Lavik E B, Qu X, Park K I, Ourednik J, 1998 Marrow stromal cells as a source of progenitor cells
Zurakowski D, Langer R and Snyder E Y 2002 Functional for nonhematopoietic tissues in transgenic mice with a
recovery following traumatic spinal cord injury mediated by phenotype of osteogenesis imperfecta Proc. Natl Acad. Sci.
a unique polymer scaffold seeded with neural stem cells USA 95 1142–7
Proc. Natl Acad. Sci. 99 3024–9 [88] Almeida-Porada G, Flake A W, Glimp H A and Zanjani E D
[72] Shihabuddin L S, Horner P J, Ray J and Gage F H 2000 Adult 1999 Cotransplantation of stroma results in enhancement of
spinal cord stem cells generate neurons after transplantation engraftment and early expression of donor hematopoietic
in the adult dentate gyrus J. Neurosci. 20 8727–35 stem cells in utero Exp. Hematol. 27 1569–75
[73] Mezey E, Key S, Vogelsang G, Szalayova I, Lange G D and [89] Beauchamp J R, Morgan J E, Pagel C N and Partridge T A
Crain B 2003 Transplanted bone marrow generates new 1999 Dynamics of myoblast transplantation reveal a discrete
neurons in human brains Proc. Natl Acad. Sci. minority of precursors with stem cell-like properties as the
100 1364–9 myogenic source J. Cell Biol. 144 1113–22
[74] Ponticiello M S, Schinagl R M, Kadiyala S and Barry F P [90] Gothot A, van der Loo J C, Clapp D W and Srour E F 1998
2000 Gelatin-based resorbable sponge as a carrier matrix Cell cycle-related changes in repopulating capacity of
for human mesenchymal stem cells in cartilage regeneration human mobilized peripheral blood CD34(+) cells in
therapy J. Biomed. Mater. Res. 52 246–55 non-obese diabetic/severe combined immunedeficient mice
[75] Solchaga L A, Gao J, Dennis J E, Awadallah A, Lundberg M, Blood 92 2641–9
Caplan A I and Goldberg V M 2002 Treatment of [91] Lazarus H M, Haynesworth S E, Gerson S L, Rosenthal N S
osteochondral defects with autologous bone marrow in a and Caplan A I 1995 Ex vivo expansion and subsequent
hyaluronan-based delivery vehicle Tissue Eng. 8 333–47 infusion of human bone marrowderived stromal progenitor
[76] Young R G, Butler D L, Weber W, Caplan A I, Gordon S L cells (mesenchymal progenitor cells): implications for
and Fink D J 1998 Use of mesenchymal stem cells in a therapeutic use Bone Marrow Transplant. 16 557–64
collagen matrix for Achilles tendon repair J. Orthop. [92] Horwitz E M et al 1999 Transplantability and therapeutic
Res. 16 406–13 effects of bone marrow-derived mesenchymal cells n
[77] Ge Z G, Goh J C H and Lee E H 2005 The effects of bone children with osteogenesis imperfecta Nature Med.
marrow-derived mesenchymal stem cells and fascia wrap 5 309–13

70
Mesenchymal stem cells for tissue engineering and regenerative medicine

[93] Fouillard L et al 2003 Engraftment of allogeneic mesenchymal press-coating biodegradable polymer with human
stem cells in the bone marrow of a patient with severe mesenchymal stem cells Tissue Eng. 8 131–44
idiopathic aplastic anemia improves stroma Leukemia [99] Tuli R, Nandi S, Li W J, Tuli S, Huang X, Manner P A,
17 474–6 Laquerriere P, Noth U, Hall D J and Tuan R S 2004 Human
[94] Dixon T, Shaw M, Ebrahim S and Dieppe P 2004 Trends mesenchymal progenitor cellbased tissue engineering of a
in hip and knee joint replacement: socioeconomic single-unit osteochondral construct Tissue Eng. 10 1169–79
inequalities and projections of need Ann. Rheum. Dis. [100] Li W J, Tuli R, Okafor C, Derfoul A, Danielson K G,
63 825–30 Hall D J and Tuan R S 2005 A three-dimensional
[95] Hunziker E B 2002 Articular cartilage repair: basic science nanofibrous scaffold for cartilage tissue engineering using
and clinical progress. A review of the current status and human mesenchymal stem cells Biomaterials 26 599–609
prospects Osteoarthritis Cartilage 10 432–63 [101] Alhadlaq A and Mao J J 2003 Tissue-engineered neogenesis
[96] Grande D A, Mason J, Light E and Dines D 2003 Stem cells as of humanshaped mandibular condyle from rat mesenchymal
platforms for delivery of genes to enhance cartilage repair J. stem cells J. Dent. Res. 82 951–6
Bone Joint Surg. Am. 85-A (Suppl. 2) 111–6 [102] Alhadlaq A et al 2004 Adult stem cell driven genesis of
[97] Fuchs J R, Hannouche D, Terada S, Vacanti J P and Fauza D O human-shaped articular condyle Ann. Biomed.
2003 Fetal tracheal augmentation with cartilage engineered Eng. 32 911–23
from bone marrow-derived mesenchymal progenitor cells J. [103] Williams C G, Kim T K, Taboas A, Malik A, Manson P and
Pediatr. Surg. 38 984–7 Elisseeff J 2003 In vitro chondrogenesis of bone
[98] Noth U, Tuli R, Osyczka A M, Danielson K G and Tuan R S marrow-derivedmesenchymal stem cells in a
2002 In vitro engineered cartilage constructs produced by photopolymerizing hydrogel Tissue Eng. 9 679–88

71

You might also like