Sunteți pe pagina 1din 12

Send Orders of Reprints at reprints@benthamscience.

net
144 Current Stem Cell Research & Therapy, 2013, 8, 144-155

Mesenchymal Stem Cells Derived from Wharton's Jelly of the Umbilical


Cord: Biological Properties and Emerging Clinical Applications

Aristea K. Batsali1,2, Maria-Christina Kastrinaki1, Helen A. Papadaki1 and


Charalampos Pontikoglou*,1

1
Department of Hematology, University of Crete School of Medicine, Heraklion, Greece; 2Graduate Program, Univer-
sity of Crete School of Medicine, Heraklion, Greece

Abstract: In recent years there seems to be an unbounded interest concerning mesenchymal stem cells (MSCs). This is
mainly attributed to their exciting characteristics including long-term ex vivo proliferation, multilineage potential and im-
munomodulatory properties. In this regard MSCs emerge as attractive candidates for various therapeutic applications.
MSCs were originally isolated from the bone marrow (BM) and this population is still considered as the gold standard for
MSC applications. Nevertheless the BM has several limitations as source of MSCs, including MSC low frequency in this
compartment, the painful isolation procedure and the decline in MSC characteristics with donor's age. Thus, there is ac-
cumulating interest in identifying alternative sources for MSCs. To this end MSCs obtained from the Wharton's Jelly (WJ)
of umbilical cords (UC) have gained much attention over the last years since they can be easily isolated, without any ethi-
cal concerns, from a tissue which is discarded after birth. Furthermore WJ-derived MSCs represent a more primitive
population than their adult counterparts, opening new perspectives for cell-based therapies. In this review we will at first
give an overview of the biology of WJ-derived UC-MSCs. Then their potential application for the treatment of cancer and
immune mediated disorders, such graft versus host disease (GVHD) and systemic lupus erythematosus (SLE) will be dis-
cussed, and finally their putative role as feeder layer for ex vivo hematopoietic stem cell (HSC) expansion will be pointed
out.

Keywords: Graft versus host disease, hematopoietic stem cells, mesenchymal stem cells, umbilical cord, Wharton's jelly.

INTRODUCTION Friedenstein et al. demonstrated that a population of non-


hematopoietic, plastic-adherent, fibroblast-like cells can also
Stem cells are defined by their unique capacity to self- be isolated from BM [3]. These cells, which were later
renew, differentiate into one or more cell lineages and tissue
shown to be able to give rise to adipocytes, chondrocytes,
types, and functionally repopulate a tissue in vivo [1]. On the
osteocytes, smooth muscle cells, fibroblasts and hema-
basis of their ability to give rise to one or more cell types,
topoietic supportive stroma [3-6] and be self-renewing [7],
known as potency, stem cells can be hierarchically classified
are currently referred to as mesenchymal stem/stromal cells
as unipotent, multipotent, pluripotent and totipotent. Unipo-
(MSCs) [4, 5, 8, 9]. So far MSC-like cells have been isolated
tent stem cells can produce only one cell type (e.g. neurons), from several tissues, such as skeletal muscle, adipose tissue,
whereas multipotent ones can differentiate into several cell
umbilical cord, synovium, dental pulp, amniotic fluid, as
types (e.g. hematopoietic stem cells, HSCs). An embryonic
well as fetal blood, liver, BM, lung and heart [10-12]. Never-
stem cell (ESC), derived from the inner cell mass (ICM) of
theless, these populations are not equivalent in terms of mul-
the embryo is pluripotent as can differentiate into cells of the
tipotency, when in vivo assays are used [13].
three germ layers, but not into trophoblastic cells, whereas
the zygote is totipotent as it produces cells of embryonic as The field of MSCs has witnessed considerable progress
well as extraembryonic tissue. over the last decade. Due to the unraveling of many of their
exceptional characteristics and the encouraging preclinical
HSCs isolated from post-natal bone marrow (BM) and
and clinical studies, it is now widely accepted that MSCs
umbilical cord blood (UCB) are generally acknowledged as
will provide a revolutionary advantage in the therapeutic
the prototype of multipotent adult tissue stem cells, being
intervention of various diseases or tissue damage in the near
able to differentiate into all mature blood lineages as well as
future. More than forty years since their initial description,
serially reconstitute hematopoiesis in lethally irradiated mice BM-MSCs represent the most extensively studied population
at the single cell level [2]. In the late 60s and early 70s,
of adult MSCs and are still considered as the gold-standard
for MSC clinical applications. Nevertheless it is now becom-
*Address correspondence to this author at the Department of Hematology, ing increasingly clear that BM may not represent the most
University Hospital of Heraklion, P.O. Box 1352, Heraklion, Crete, Greece; suitable source for MSC collection. Besides the invasive and
Tel: +30 2810 394629 ; Fax: +30 2810 394632 ; painful isolation procedure, MSCs are present at very low
E-mail: pontikoglou@yahoo.com frequency (approximately 0.001–0.01% of BM mononuclear

2212-3946/13 $58.00+.00 © 2013 Bentham Science Publishers


Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 145

cells [14]) in BM-aspirates and in addition their qualitative Human Umbilical Cord is a Rich Source of MSCs
and quantitative properties decline with donor's age [15].
The human UC (Fig. 1) contains two arteries and a vein
The low frequency implies that an extensive in vitro expan-
buried within a mucous or gelatinous connective tissue also
sion of cells is usually required to obtain sufficient cell num-
known as the Wharton’s jelly (WJ), enclosed by a simple
bers for therapeutic interventions, thereby enhancing the risk
of epigenetic damages as well as viral and bacterial contami- amniotic epithelium. WJ is largely made from proteoglycans
and various types of collagen, forming a sponge-like tissue,
nations. To circumvent these obstacles, much effort has been
within which stromal cells are embedded [19]. These cells,
invested over the last decade in identifying alternative, more
also referred to as umbilical cord matrix (UCM) cells are
abundant and easily attainable sources of MSCs for thera-
thought to be trapped into the WJ early in embryogenesis
peutic use. In this regard the umbilical cord (UC), which is
during their migration from the aortic-gonadotropin-
considered medical waste and is obtained with a painless,
simple and safe procedure during delivery, has aroused much mesonephric region to the fetal liver through the umbilical
cord [20]. UCM cells have exceptional properties in that
attention during the last years as a promising source for MSC
although they are bona fide MSCs [18, 21] as discussed be-
isolation. MSCs have actually been found in both UC blood
low in detail, possessing similar properties with their adult
(UCB) as well as in UC tissues. Nevertheless, the scarcity of
BM counterparts, they also retain characteristics of primitive
MSC in UCB (their frequency being much lower than in
stem cells, like the expression of embryonic stem cell mark-
BM) and the lack of a robust and reproducible technique for
harvesting and expanding MSCs from UCB units [16, 17] ers [22, 23]. In line with these observations MSCs derived
from WJ, hereafter referred to as "UC-MSCs", maintain their
have hampered their use in clinical applications. On the other
self-renewal capacity in vitro for a longer period of time, and
hand existing isolation protocols have constantly induced the
exhibit a shorter doubling time and a broader pluripotency as
harvest of large numbers of MSCs from UC-tissues with a
compared to BM-MSCs [18]. These unique developmental
hundred per cent success.
and operational characteristics along with the fact that UC-
UC-MSCs share many properties with adult BM-MSCs, MSCs can be easily isolated, extracorporeally without risks
but are generally considered as a more primitive population for the donor, from a tissue which is routinely discarded,
than the latter. In fact, while they possess several characteris- highlight the advantages of UC-MSCs over ESCs and adult
tics of ESCs, they do not form teratomas upon transplanta- MSCs and explain the rapidly growing interest in this field
tion [18] and their research does not raise ethical or legal of stem cell biology.
issues. All these features open attractive perspectives for the
The first report providing robust evidence that WJ stro-
propagation and engineering of UC-derived MSCs for cell-
based therapies. This review will first attempt to give a mal cells can be classified as MSCs was published in 2004
[24]. In that study, cells extracted from WJ, displayed a fi-
summary of the biology of MSCs derived from Wharton's
broblast-like phenotype when expanded in vitro and were
Jelly (WJ-MSCs), the most abundant region of UC and then
able to differentiate into osteoblasts, adipocytes and chon-
discuss their potential application for the treatment of cancer
drocytes upon appropriate stimulation [24]. Furthermore,
and immune mediated disorders such as graft versus host
akin to BM-MSCs they were shown to express high levels of
disease (GVHD) and systemic lupus erythematosus (SLE).
Finally, their putative role as feeder layers for ex vivo hema- CD29, CD44, CD51, CD73 and CD105, but lacked expres-
sion of the hematopoietic markers CD34 and CD45 (Table 1)
topoietic stem cell (HSC) expansion will be pointed out.
[24]. Subsequent reports have provided additional informa-

Umbilical arteries
Perivascular

Intervascular
Wharton’s Jelly

A
Amniotic epithelium Umbilical vein/ Subamnion
Umbilical cord blood
Fig. (1). Umbilical cord contains two arteries and a vein, within a mucous or gelatinous connective tissue, the Wharton’s jelly, enclosed by a
simple amniotic epithelium. Mesenchymal stem cells have been isolated from umbilical cord blood and also from the intervascular, perivas-
cular and subamniotic regions of Wharton's jelly.
146 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.

tion on WJ stromal cell phenotype, demonstrating the ex- remove umbilical cord vessels, scrape off WJ with a scalpel
pression of various membrane molecules known to be asso- and then treat the tissue enzymatically [24]. Others cut down
ciated with a MSC-like identity. These molecules include UC to smaller segments, strip the vessels and then directly
CD90, CD146, CD166 and ganglioside 2 (GD2) [8, 23, 25, immerse the remaining UC to an enzymatic solution [25, 27,
26]. Collectively, the morphology, immunophenotype and 31]. Interestingly, neither the enzymes used nor the incuba-
differentiation potential of WJ stromal cells fulfill the criteria tion period have been standardized thus far. Alternatively,
for MSCs proposed by the International Society for Cellular instead of enzymatic digestion, several groups dissect the
Therapy (ISCT) [8]. Indeed, given that at present there is no cord segment into very small pieces, with or without discard-
specific marker to define UC-MSCs, the expression of the ing the cord vessels and the segments are used as explants
aforementioned set of membrane antigens combined with the from which cultures are subsequently established [32-34].
demonstration of in vitro multi-lineage differentiation poten- The major advantage of this method is that it minimizes cell
tial is necessary to identify MSCs in UC-derived tissues damage as it does not involve enzymatic treatment, however
it should be noted that primary cultures generated in this way
UC-MSCs have been isolated from three relatively indis-
are quite heterogeneous [33]. UC-MSCs have also been iso-
tinct regions of the WJ: the perivascular, the intervascular
lated from WJ by simply cutting UC segments longitudinally
and the subamnion zones (Fig. 1) [27]. Cells from these
and plating them with WJ side-down onto a plastic surface,
zones share a fibroblast-like morphology, a high proliferative
once again without enzyme digestion or dissection [35]. Fi-
rate and multipotent differentiation capacity. However a nally, to collect HUCPVCs some groups remove blood ves-
comprehensive head-to-head comparison between MSCs
sels together with surrounding WJ, tie up the ends of each
obtained from different regions of the UC has not been per-
vessel into a loop and then immerse the vessel in enzyme,
formed thus far, due to the great variation in cell isola-
thereby isolating cells from the perivascular tissue [28, 29].
tion/culture protocols. Hence the question whether cells har-
vested from different components of the UC represent differ- The question as to which is the optimal method for effi-
ent populations is still open. Nevertheless one study combin- cient isolation of a homogeneous and well defined popula-
ing both in situ and in vitro experiments [27] has demon- tion of UC-MSCs has not been answered thus far. However,
strated that MSCs derived from the perivascular region com- unless an agreement is reached between laboratories regard-
prise a distinct subpopulation than those derived from the ing the isolation and ex vivo expansion of UC-MSCs, there
intervascular and subamniotic parts of the UC. It has thus will be no consensus on the characteristics and biological
been reported that perivascular MSCs stain strongly for pan- properties of the cells, which is a prerequisite for the proper
cytokeratin as compared to intervascular and subamniotic design of preclinical studies, so as to pave the way for the
counterparts and give rise in vitro to flat, wide cytoplasmic potential use of UC-MSCs in clinical applications.
cells, named type 1 cells, which tend to diminish in number
during protracted culture [27]. On the other hand intervascu- Do differences in isolation procedures and ex vivo expan-
lar and subamniotic MSCs generate fibroblast-like cells per- sion protocols have an impact on the antigens expressed by
sisting throughout culture (type 2 cells). In view of these UC-MSCs? This issue has not been properly investigated so
findings it appears that UC-MSCs are actually type 2 cells, far, yet a recent study [36] demonstrated that MSC markers
displaying adipogenic, chondrogenic, osteogenic and neu- CD73, CD90 and CD105 varied their expression patterns
ronal capacity, upon appropriate differentiation stimuli [27]. according to whether enzyme cocktails or explants were used
Isolation of MSCs from UC perivascular zone has been re- for UC-MSC harvesting. A similar variation with respect to
ported by other groups as well [28, 29]. These cells, termed the isolation method was also noted in the expression of the
as Human Umbilical Cord Perivascular Cells (HUCPVCs), ESC markers c-Kit and Oct-4 by UC-MSCs [36].
have a colony forming unit (CFU) frequency of 1:333 [29]
and differ in their growth characteristics from UC-MSCs WJ-derived UC-MSCs may be Regarded as a Primitive
(population doubling times ranging from 59.4±42.4h in pas- Stromal Cell Population
sage 0 to nearly 20h in passage 6 for HUCPVCs versus
85±7.2h in passage 0 to 11±1.2h in passage 7 for a mixed Porcine UC-MSCs have been reported to express the
population of UC-MSCs [27, 29]. Furthermore, HUCPVCs ESC markers Oct-4, Sox-2 and Nanog [37] and equine UC-
exhibit higher proliferative capacity and osteogenic potential MSCs express, apart from Oct-4, the embryonic markers
as compared to BM-MSCs [28]. The phenotype of SSEA4 and c-Kit [38]. In line with these findings, human
HUCPVCs shares many similarities with that of WJ-derived UC-MSCs express Oct-4, Nanog, Sox-2, c-Kit, as well as
MSCs (Table 1). One significant difference though, is the Dnmt3b and Tert, albeit at lower levels as compared to ESCs
high expression of CD146 by HUCPVCs. Interestingly this [22, 34, 39]. Such a low expression of the aforementioned
marker is either absent [28] or weakly expressed by MSCs pluripotency markers may explain why UC-MSCs do not
derived form other regions of WJ [30]. produce teratomas in vivo in immunodeficient mice and rats
[22, 25]. In addition human UC-MSCs express the ESC
Isolation of MSCs from Wharton's Jelly
markers Tra-1-60, Tra-1-81, SSEA-1 and SSEA-4 [22].
In recent years, several methods (Fig. 2) have been de- Many of these pluripotent stem cell markers retain their ex-
veloped for the isolation of MSCs from WJ. However, ex- pression for at least nine passages during ex vivo expansion.
perimental approaches vary considerably and in some cases Comparative gene expression profiling between UC-MSCs
harvested cells from UC tissue most likely consist of a het- and BM-MSCs demonstrated that the former express higher
erogeneous population, thereby biasing comparisons. With levels of Nanog, Dnmt3b and Gabrb3, and also
regards to protocols used to isolate UC-MSCs, some authors
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 147

Table 1. Markers expressed by BM-MSCs and UC-MSCs derived from WJ and the perivascular zone

Markers WJ-derived MSCs Perivascular MSCs BM-MSCs

CD10 neutral endopeptidase + [25] + [103] + [101]


CD11b Integrin alpha-M - [23] - [102]
CD13 aminopeptidase N + [25] + [102]
CD14 LPS receptor - [25] - [49] - [102]
CD26 dipeptidylpeptidase 4 + [102]
CD29 intergrin 1 + [25] + [49] + [5]
CD31 PECAM-1 (platelet endothelial adhesion molecule-1) - [25] - [103] - [5]
CD33 sialic acid binding Ig-like lectin 3 - [81] - [101]
CD34 sialoprotein - [24] - [29] - [5]
CD38 ADP-ribosyl cyclase 1 - [47] - [101]
CD40 tumor-necrosis factor receptor superfamily 5 - [61] + [101]
CD44 hyaluronan receptor + [25] + [29] + [5]
CD45 pan-leucocyte antigen - [25] - [29] - [5]
CD49a integrin 1 - [104] + [5]
CD49b intergrin 2 + [25] + [5]
CD49c intergrin 4 + [25] + [5]
CD49d intergrin 3 + [25] - [5]
CD49e integrin 5 + [25] + [5]
CD50 ICAM-3 (Intercellular adhesion molecule 3) - [105] - [5]
CD51 integrin V + [25] + [5]
CD53 Tetraspanin-25 - [104] - [102]
- [47]
CD54 ICAM-1 (intercellular adhesion molecule-1) + [29] + [5]
+ [105]
CD56 NCAM-1 (neural cell adhesion molecule) - [61] - [5]
CD58 LFA-3 (lymphocyte function-associated antigen 3) + [81] + [101]
CD71 transferin receptor - [106] + [100]
CD73 Ecto-5 nucleotidase + [104] + [29] + [5]
CD80 CD28 ligand - [81] - [102]
CD86 Activation B7-2 antigen - [81] - [102]
CD90 Thy-1 + [25] + [29] + [5]
CD105 TGF-RIII (transforming growth factor- receptor III) + [25] + [29] + [5]
- [49] - [29]
CD106 VCAM-1 (Vascular cell adhesion molecule-1) + [5]
+ [61] + [107]
- [104] - [49]
CD117 C-kit receptor - [102]
+ [108] + [29]
CD133 AC133 (prominin) - [25] - [109]
CD140 PDGF-R (platelet-derived growth factor receptor ) - [54] + [102]
CD140 PDGF-R (platelet-derived growth factor receptor ) ND + [102]
CD144 VE-cadherin Low [30] - [5]
CD146 Mel-CAM (melanoma-cell adhesion molecule) Weak + [30] + [49] + [7]
148 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.

Table 1. Contd….

Markers WJ-derived MSCs Perivascular MSCs BM-MSCs

CD166 ALCAM (activated lymphocyte cell adhesion molecule) + [104] + [5]


CD235 Glyocophorin - [29] - [102]
CD325 N-cadherin + [25] + [102]
HLA-I + [54] + [29] + [110]
HLA- II - [54] - [29] - [110]
HLA G + [34] + [111]
- [81]
STRO-1 + [113]
+ [112]
SSEA4 Stage-specific embryonic antigen 4 + [105] + [114]
a-SM actin a-smooth muscle actin + [54] - [103] + [10]
NANOG + [25] + [115]
OCT4 + [34] - [29] + [115]
SOX2 + [25] - [6]
GD2 Ganglioside + [26] + [10]

Umbilical cord

Removal of umbilical
Dissection in vessels
small segments

Scrapping off
mesenchymal tissue
Cut tissue into
very small pieces

Enzymatic treatment

Explant culture Filter

Stromal cell culture

Fig. (2). A scheme depicting the different approaches followed for the establishment of WJ-derived UC-MSC cultures (modified from [100]).
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 149

show an increased expression of the pluripotent/stem cell finitive evidence that UC-MSC-derived cardiomyocytes rep-
markers Brix, CD9, Gal, Kit and Rex1 [39]. The implication resent functional and terminally differentiated cells is still
of these markers in the biology of UC-MSCs is currently scarce. To our knowledge, only one group has thus far re-
unknown, yet their expression is thought to reflect the more ported that UC-MSC-derived cardiomyocytes exhibit slight
primitive nature of UC-MSCs as compared to their adult spontaneous beating after 21 days of induction [31].
counterparts within the BM. However, not all UC-derived
Finally, several groups have shown that UC-MSCs can
MSCs express ESC markers. In fact, it has been demon-
acquire morphological and biochemical properties of neural
strated that primitive markers are expressed only by a subset
cells when exposed to appropriate differentiation media [27,
of UC-MSCs [26, 40], thereby suggesting that these cells are
32, 51, 52]. Mitchell et al. [32] were the first who observed
rather hetereogeneous in terms of stemness. It would there-
UC-MSC neuronal differentiation, following a complex and
fore be very challenging to identify that specific marker
multi-step neuronal induction process. Differentiation was
which would permit the isolation of the primi-
assessed by changes in morphology and expression of neu-
tive/multipotent MSCs from UC.
ron specific enolase, bIII-tubulin, neurofilament and tyrosin
Human UC-MSCs exhibit also higher expression of hydroxylase. Subsequent studies have further corroborated
genes implicated in the phosphoinositide 3-kinase (PI3K)- these findings. It has also been suggested that UC-MSCs
AKT survival/proliferation pathway, including GAB2, may be more primed for neuronal differentiation as com-
NFKBIA, FOXO1A, YWHAB, FOXO3A, CDKN1A, pared to BM-MSCs, since they express, higher levels of neu-
MAP3K8, NFKB1, NOS3, DDR1 and TEK [41]. Further- ronal-associated genes including NESTIN, GFAP and
more, the NF-B pathway has been reported to be more ac- SEM3A [39] in a non–differentiated state. However, it
tive in UC-MSCs than in BM-MSCs [41]. These findings should be kept in mind that definitive evidence for electro-
together with the fact that human WJ-MSCs express the physiological activity of UC-MSC-derived neurons is still
primitive stem cell marker TERT [25] explain, at least in missing. Therefore the issue whether the neuronal-like phe-
part the observation that UC-MSCs have a shorter doubling notype of UC-MSCs reflects a genuine trans-differentiation
time [27] and a more extensive ex vivo expansion capacity is still pending.
[18]. Recent studies suggest that UC-MSCs can also differen-
Multilineage in vitro Differentiation Potential of WJ- tiate into endodermal lineages (reviewed in [53]). In this
derived UC-MSCs context, Campard et al. [54] reported that non-differentiated
UCM cells constitutively expressed markers of hepatic line-
Accumulating evidence suggests that UC-MSCs can be age, such as albumin, alpha-fetoprotein, cytokeratin-19, con-
successfully induced to differentiate into mesodermal and nexin-32, and dipeptidyl peptidase IV. After in vitro hepatic
non-mesodermal lineages. As mentioned above, several induction, cells exhibited hepatocyte-like morphology and
groups have established that UC-MSCs can generate os- upregulated the expression of certain hepatocyte-associated
teoblasts, adipocytes and chondrocytes in vitro [24, 27, 42, markers, but lacked important functional properties of ma-
43], thereby fulfilling one of the minimal criteria proposed ture liver cells, thereby questioning whether UCM cells have
by ISCT for MSCs [8]. However, lipid accumulation takes actually undergone hepatocyte maturation [54]. Other groups
much longer in UC-MSCs and the adipocytes are less mature have also claimed that UC-MSCs can be induced to form
than those derived from BM-MSCs [27, 41]. In addition, isle-like cells [55, 56] (reviewed in [57]), able to express
UC-MSCs have been shown to possess weaker osteogenic pancreatic associated genes and release insulin in response to
[41] and chondrogenic [44, 45] potential as compared to physiological glucose levels [56]. Interestingly, UC-MSCs
BM-MSCs. have been reported to have a higher pancreatic differentia-
UC-MSCs have also been reported to successfully differ- tion potential than their BM counterparts [55].
entiate into endothelial cells [43, 46] and actually demon- The Immunomodulatory Properties of UC-MSCs
strate higher endothelial differentiation potential when com-
pared to BM-MSCs [43]. In accordance with this observation Apart from the multilineage potential, UC-MSCs possess
non-differentiated UC-MSCs exhibit higher expression of various immunoregulatory properties, rendering them as
endothelial associated genes including FLT1, GATA4, appealing candidates for cell based therapies [58]. More pre-
GATA6, ISL1, LAMA1, SOX17 and SERPINA1 [39]. cisely UC-MSCs have been found to exhibit very low ex-
pression of HLA class I, and lack the expression of HLA-DR
One group has suggested, that UC-MSCs can give rise to [59, 60]. Notably, in comparison to BM-MSCs derived from
skeletal muscle cells when cultured in the appropriate me- aged donors, UC-MSCs express significantly lower levels of
dium and that non-differentiated UC-MSCs may display HLA-I, and exhibit significantly higher intracellular concen-
myogenic potential in vivo [47]. Differentiation of UC-MSCs trations of the immunosuppressive molecule HLA-G [59].
to cardiomyocytes has also been reported but is still contro- These findings together with the observations that UC-MSCs
versial [24, 31, 48-50]. For instance, one group [24] demon- do not express the co-stimulatory molecules CD40, CD80
strated that UC-MSCs could be appropriately induced to and CD86 [59, s61], and do not elicit proliferative responses
form cells demonstrating cardiomyocyte morphology and from allogeneic T cells in vitro [61], suggest that UC-MSCs
expressing cardiac markers, including N-cadherin and car- may be characterized by inherently low immunogenicity.
diac troponin. Another team [49], however, was unable to However, another study suggests that UC-MSCs may not be
detect cardiomyocyte marker expression after in vitro induc- as immune-privileged as thought, as they can trigger weak
tion of MSCs isolated from WJ and perivascular tissues. De- allogeneic immune activation in vitro and will eventually
150 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.

undergo rejection following xenogeneic transplantation [59]. transduction pathways are implicated in the regulation of B
Similar findings have been reported for BM-MSCs as well. cell proliferation and differentiation [67].
Interestingly the latter have been shown to be much more
The immunoregulatory potential of HUCPVCs has also
immunocompetent compared to UC-MSCs [59].
been assessed in a recent study [68]. More precisely it has
Several studies have been published on the immunosup- been demonstrated that when HUCPVCs are co-cultured
pressive potential of UC-MSCs on T lymphocytes [59, 60, with resting or stimulated allogeneic lymphocytes they fail to
62-64]. One group reported that UC-MSCs target CD4+ and induce their proliferation. Furthermore, HUCPVCs have
CD8+ T subpopulations equally and are able to inhibit T-cell been found to reduce in vitro alloreactivity in mixed lym-
proliferation regardless of the stimulus used to activate T- phocyte reactions. Notably, the immunosuppressive effect of
cells [63]. Furthermore, UC-MSC-mediated inhibitory ef- the cells is exerted, at least in part via soluble factors [68].
fects were higher than with BM-MSCs. In accordance with
previous studies suggesting that the immunomodulatory ef- UC-MSCs as Feeder Layers for ex vivo Umbilical Cord
Blood Expansion
fect of MSCs on T cells may be mediated, at least in part, by
soluble factors (reviewed in [65]), such as hepatocyte growth Despite the fact that BM has been the most extensively
factor (HGF), prostaglandin E2 (PGE2), transforming growth studied source for HSC transplantation, the importance of
factor-1 (TGF-1) and indoleamine 2,3-dioxygenase (IDO), UCB as an alternative source of transplantable HSCs has
the authors assessed whether any of these molecules may be been widely acknowledged since the first successful UCB
implicated in UC-MSC immunoregulation as well. In their transplant in 1988 [69]. From a practical point of view UCB
hands immune suppression exerted by both UC- and BM- transplantation has several advantages over BM, including
MSCs was mediated by cycloxygenase (COX) 1 and COX2 the non-invasiveness of the procedure, the ease of procure-
enzymes, regulating PGE2 synthesis, and by the production ment, the reduced risk of transmissible infections and the
of PGE2 per se, whereas HGF did not seem to be actively availability for immediate use [70]. In addition, UCB grafts
implicated. These findings were further corroborated by an- allow for a greater tolerable HLA disparity with a decreased
other study [64] which additionally showed that while UC- frequency of acute or chronic graft versus host disease
MSCs intrinsically secrete very low levels of PGE2, its pro- (GVHD) as compared to BM grafts [70]. Finally UCB con-
duction is upregulated when cells are co-cultured in the pres- tains a higher proportion of immature and committed hema-
ence of activated human peripheral blood mononuclear cells topoietic progenitor cells (HPCs) in comparison to adult BM
(hPBMCs). The authors attributed the PGE2 stimulation to [71].
inflammatory cytokines (including IFN- and interleukin-1)
produced by hPBMCs [64]. However UCB transplantation has also some significant
restrictions, limiting its broad applicability. The most sig-
Interestingly, several studies [59, 62, 66] have reported nificant limiting factor of UCB use is probably the low total
that UC-MSC immune characteristics may be modified fol- cell dose harvested from a UCB unit, which in turn is associ-
lowing exposure to inflammatory cytokines and/or activated ated with delayed neutrophil and platelet engraftment and
PBMCs/T-cells so as to display immunosupressive proper- higher engraftment failure [72]. For this reason UCB trans-
ties, a process described by the term licensing or priming. In plantation remains significantly more successful in children
this context, Deuse et al. [59] reported that IFN- at doses [73], whereas poor outcomes have been reported in patients
below 50ng/ml upregulated HLA-DR and HLA-I expression, >45 kgr who receive only one unit of UCB [74].
thereby increasing UC-MSC immunogenicity. IFN- treat-
ment also induced tolerogenicity by increasing intracellular To circumvent these obstacles, so as to improve out-
HLA-G expression, TGF- and IL-10 release, and stimulated comes and extend the application of UCB transplantation to
the production of IDO [59]. Higher doses of IFN- however older and/or larger patients, several groups have explored the
enhanced UC-MSC immunosuppressive phenotype by possibility of in vivo expansion of HSCs from UCB units.
downregulating HLA-DR expression and further increasing Various protocols are under investigation over the last 30
IDO. These results suggest that appropriate priming may be years, including those based on cultures on feeder layers.
crucial for the fine adjustment of MSC characteristics, ren- Among the different cell types that have been used as stro-
dering them suitable for a given therapeutic application. mal support for UCB expansion, the most extensively stud-
ied thus far are BM- MSCs.
The immunomodulatory effects of MSCs are not only
apparent in T lymphocytes but also concern B lymphocytes. The rationale behind the use of MSCs as feeder cells for
To our knowledge however, the impact of UC-MSCs on B UCB expansion lies on a large body of evidence, clearly
cells has been poorly investigated and undoubtedly awaits demonstrating the BM-MSC promoting role on hematopoie-
further elucidation, as the information that will be gathered is sis (reviewed in [65]). More precisely BM-MSCs have been
expected to provide crucial clues for the introduction of WJ- shown in vivo to enhance HSC engraftment and repopula-
MSCs into the clinic. In the single study reported until today tion, as evidenced by the observation that co-transplantation
[67] UC-MSCs were found to inhibit B cell proliferation, of human HSCs and MSCs results in increased chimerism
differentiation and antibody production in vitro. This sup- and/or hematopoietic recovery, in both animal models and
pressive effect is mediated, at least in part by soluble factors, humans [65, 75]. On the other hand, MSCs have been shown
which may downregulate Blimp-1 and upregualate PAX-5, in vitro to maintain long-term culture initiating cells (LT-
two master regulators of B cell differentiation [67]. Further- CICs) and expand lineage-specific colony forming units
more UC-MSCs have been demonstrated to inhibit Akt and from CD34+ marrow cells in long-term BM cultures (re-
p38 MAPK phosphorylation. Interestingly, these signal viewed in [65]). Even though the exact mechanisms that me-
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 151

diate the hematopoiesis promoting effect of MSCs have not support the growth and maintenance of UCB hematopoietic
been elucidated, it is now widely accepted that MSCs supply stem/progenitor cells in long term culture [82, 83], thereby
HSCs and their progeny with signals for survival, prolifera- establishing an ex vivo model that could enable the study of
tion and differentiation through direct cell-to-cell contact the interactions between HSCs and their supportive micro-
and/or production of cytokines, adhesion molecules and ex- environment within the UC. However, whether UC MSC-
tracellular matrix proteins (reviewed in [65]). based HSC expansion will have possible future clinical util-
These observations have provided the theoretical back- ity is currently unknown and awaits further clarification.
ground for the investigation of the potential use of MSCs as
scaffolds for the ex vivo expansion of HSCs. Based on the UC-MSCs for GVHD Treatment
hypothesis that MSCs could provide many of the molecular Graft versus host disease (GVHD) is one of the most
cues of the hematopoietic microenvironment necessary to severe complications of allogeneic BM transplantation. First-
favor HSC self-renewal and regulate their proliferation, CB line treatment with corticosteroids is the gold standard for
hematopoetic stem/progenitor cells have been co-cultured on this T-cell mediated process, however second line treatment
BM-MSC preformed layers in a medium containing (FBS) for steroid-resistant GVHD has not been clearly established
and various combinations of growth factors including G- thus far and the outcome is poor, with one year overall sur-
CSF, stem cell factor (SCF), thrombopoietin (TPO), Flt3- vival (OS) of only 30% [84, 85].
ligand or fibroblast growth factor-1 (FGF-1) [76]. Using this
The in vitro and in vivo immunomodulatory properties of
ex vivo model several groups have demonstrated an increase
BM-MSCs have provided the theoretical background for the
in the number of total nucleated cells (TNC), clonogenic
use of these cells for treatment of acute steroid-resistant
cells or CD34+ cells [77, 78]. These findings were further GVHD. Several reports have been published thus far (re-
corroborated in a recent study [79], where CB CD34+ cells viewed in [86]) suggesting that clinical responses can actu-
were co-cultured with BM-MSCs in a serum free medium ally be attained in a significant number of patients, especially
supplemented with SCF, TPO and FGF-1. BM-MSCs were in children. BM-MSCs have also been used for the treatment
also shown to maintain the primitive phenotype of CB hema- of chronic GVHD showing likewise promising results [86].
topoietic stem and progenitor cells even after massive expan-
sion [79]. However co-culture with BM-MSCs did not result To our knowledge, there has been only one recent study
in a significant expansion of long-term repopulating cells. A clinically testing UC- MSCs for GVHD [87]. In that particu-
clinical trial is underway at M.D. Anderson Cancer Center lar report third-party culture expanded UC- MSCs were
combining an unmanipulated UCB unit with an expanded transplanted in 2 children with severe steroid-resistant acute
UCB unit for 14 days in the presence of a cytokine cocktail GVHD. The first patient received three infusions of UC-
on layer of family member donor-derived BM-MSCs or "off- MSCs. Each time acute GVHD improved significantly and
the-self" BM-MSCs (Angioblast Systems Inc.). Preliminary eventually disappeared. The second patient received only
results suggest a beneficial role for these stromal cell-based one infusion of UC-MSCs that led to complete remission of
HSC expansion [80]. acute GVHD. No child experienced any adverse events what
so ever and both children were well and remained off immu-
Given the promising role of BM-MSCs in supporting ex nosuppressive therapy at the time the study was published.
vivo UCB expansion and since UC-MSCs share many char- The authors concluded that UC-MSCs may be considered as
acteristics with their BM counterparts, but in contrast to the a feasible, safe and effective treatment modality for acute
latter can be readily and painlessly harvested from UC, they GVHD [87]. These encouraging results need obviously to be
soon emerged as attractive candidates for HSC expansion. In confirmed by larger trials, which will include adult patients
this regard, the first report demonstrating that akin to BM- as well. Furthermore, additional studies are needed in order
MSCs, UC-MSCs can also support hematopoiesis was pub- to optimize the use of UC-MSCs in treating GVHD. For ex-
lished in 2007 [81]. In that study the authors showed that ample, it is currently unknown which is the optimal UC-
UC-MSCs secrete significantly higher amounts of several MSC dose and schedule, or whether the use of platelet-
important cytokines and hematopoietic growth factors in- lysate, or other substitutes of the standard FBS-enriched me-
cluding G-CSF, GM-CSF, LIF, IL-1, L-6, IL-8 and IL-11, dium (so as to comply with regulatory guidelines for the
compared to BM-MSCs. Furthemore co-culture of UC- production of MSCs for clinical use, which recommend
MSCs with UCB CD34+ cells significantly increased hema- avoiding the use of animal derivatives) might influence the
topoietic colony formation as compared to clonogenic pro- therapeutic effect of UC-MSCs.
genitor cell assays performed in the absence of MSCs [81].
Finally, in agreement with previous reports demonstrating UC-MSCs Support the Expansion of Natural Killer Cells
that co-transplantation of BM-MSCs with HSCs can acceler- As previously mentioned a major toxicity associated with
ate hematopoietic engraftment, co-injection of UC-MSCs Bone Marrow Transplantation (BMT) is GVHD, an attack
with UCB cells or CD34+ cells in a NOD/SCID mouse by the T cells in the graft against the recipient organs includ-
model accelerated human HSC recovery. The authors as- ing the skin, gastrointestinal tract, liver, and eyes [88]. Ag-
sumed that the improved engraftment might be attributed to gressive depletion of T cells in the allogeneic graft can
the presence of UC- MSCs within the BM of the recipient markedly reduce the incidence of acute GVHD, however,
animals and also to the production of hematopoietic cytoki- this is associated with significant drawbacks, including re-
nes by these cells. Subsequent reports have confirmed the duction of the graft versus tumor effect of the allogeneic-
capacity of UC-MSCs and UC-MSC conditioned medium to BMT [89]. Limiting therefore GVHD while maintaining the
152 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.

antitumor effect, would be critical to improve transplant out- other tissues. This combined approach resulted in significant
come [90]. Given that natural killer (NK) cells have been metastatic tumor reduction, much greater than compared to
found to control leukemia relapse while protecting patients each treatment alone. A more recent study has corroborated
from GVHD [91, 92], the possibility of expanding and po- the ability of UC-MSCs to reduce the growth of human
tentially using these cells for adoptive immunotherapy ap- breast carcinoma cells both in vitro as well as in vivo, in a
pears an attractive approach in the setting of allogeneic mouse xenograft model [98]. Notably, the antitumor proper-
BMT. Provided that UCB is rich in NK cell progenitors and ties of UC-MSCs do not seem to be specific only to breast
since UC-MSCs can produce significant amounts of cytoki- cancer cells, but have also been demonstrated in osteosar-
nes, Boissel et al. [93] hypothesized that a feeder layer of coma and ovarian carcinoma cell lines in vitro [99]. The
UC-MSCs might actually effectively support ex vivo expan- mechanisms mediating UC-MSC anticancer effects are still
sion of UCB-NK cells. Indeed when CD3-depleted UCB under investigation. However, it has been suggested that UC-
mononuclear cells were cocultured with UC-MSCs and a MSCs exert their antitumor activity both via cell-to-cell con-
cytokine cocktail, the proportion of CD3-CD56+ NK cells tact and humoral factors, through induction of apoptosis, cell
increased significantly as compared to cell cultured with cycle arrest and autophagy [97, 99]. Taken together, the
cytokines only. Interestingly direct contact between UCB aforementioned preliminary data imply that UC-MSCs may
mononuclear cells and UC-MSCs resulted in a significantly hold promise as a therapeutic modality in cancer and high-
greater NK cell expansion. Notably, expanded NK cells light the need for the development of additional well-
maintained an elevated cytotoxic profile [93]. The possibility designed pre-clinical studies so as to properly investigate the
of further optimizing this experimental protocol should be potential of this cell-based approach.
evaluated in additional studies and of course the cytotoxicity
of the expanded NK cells awaits assessment in animal mod- CONCLUSION
els.
Within recent years considerable interest has been given
UC-MSC Transplantation in Systemic Lupus Erythema- to the exciting properties of UC-MSCs, derived from the WJ,
tosus because of their primitive nature as compared to BM-MSCs
(the prototype of adult MSCs), their multilineage potential
To our knowledge, apart from GVHD, in vivo therapeutic
across germ layers, the extensive in vitro proliferation and
efficacy of UC- MSC transplantation has also been clearly
their immunosuppressive capacity. These characteristics
illustrated in the setting of systemic lupus erythematosus
along with the ease by which UC-MSCs are isolated from a
(SLE). The theoretical background for assessing the clinical
tissue with unlimited availability, have generated much en-
impact of UC-MSCs in patients with this autoimmune disor-
der has been provided by the immunomodulatory properties thusiasm regarding their potential applications in cell-based
therapies. This expectation is being supported by both pre-
of UC-MSCs and also by the beneficial effects of BM- MSC
and clinical data. In particular, in this review we reported on
administration in patients with treatment-refractory SLE
the promising results obtained in recent studies regarding the
[94]. In this context, it has recently been reported that trans-
use of UC-MSCs for the treatment of cancer and immune
plantation of UC-MSCs in patients with severe and refrac-
mediated disorders, such as graft versus host disease and
tory SLE results in clinical and serologic improvement and is
not associated with mortality or other treatment-related ad- systemic lupus erythematosus and discussed UC-MSC role
as feeder layers for ex vivo HSC expansion. Nevertheless
verse events [95]. Even though the mechanisms mediating
much work has to be done so as to fully unravel the biologic
the therapeutic effect of UC-MSCs in SLE patients remain
features of UC-MSCs and especially those distinguishing
unclear, it has been suggested that disease remission may be
them from adult MSCs. Furthermore, properly designed
associated with an increase in the proportion of Tregs in pe-
comparative studies need to be undertaken in the near future
ripheral blood along with a decrease in IL-4 serum levels,
probably resulting in inhibition of humoral activity [95]. so as to clearly identify and characterize MSC subpopula-
tions originating from different regions of WJ, in order to
Another recent study has demonstrated that UC-MSCs can
answer the long-standing question: whether different ana-
also provide clinical benefit in diffuse alveolar hemorrhage,
tomical locations within WJ are associated with distinct
a rare complication of SLE with high clinical mortality [96].
qualitative and quantitative MSC properties. Notably, avail-
These findings suggest that UC-MSC transplantation may
able global expression technologies, like gene expression
hold promise as a treatment modality in selected SLE cases.
However, the efficacy and safety of this strategy need to be and comparative genomic hybridization (CGH) microarrays,
proteome and secretome profiles, methylation mapping and
further validated in larger clinical trials with longer periods
ChIP-on-ChiP technologies are anticipated to contribute sig-
of follow-up.
nificantly to the better understanding of UC-MSC biology. It
Therapeutic Potential of UC-MSCs in Cancer is anticipated that the data obtained by these studies will not
only provide the theoretical background for the eventual in-
An emerging clinical application of UC-MSCs that re-
troduction of UC-MSCs in the clinic, but would also be ma-
cently gained much attention is their use as cellular therapy jor contribution to stem cell biology in general.
to combat cancer. One group has thus administered UC-
MSCs engineered to secrete interferon-, in combination
with low-dose intraperitoneal injections of 5-fluorouracil in CONFLICT OF INTEREST
severe combined immunodeficiency (SCID) mice, bearing The authors confirm that this article content has no con-
human breast carcinoma tumors in their lungs [97]. Inter- flicts of interest.
estingly, UC-MSCs were only found in the lungs and not in
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 153

ACKNOWLEDGEMENTS from human Wharton's jelly. Reprod Biomed Online 2007; 15:
708-18.
Declared none. [23] Fong CY, Chak LL, Biswas A,et al. Human Wharton's jelly stem
cells have unique transcriptome profiles compared to human
REFERENCES embryonic stem cells and other mesenchymal stem cells. Stem Cell
Rev 20011; 7: 1-16.
[1] Weissman IL, Anderson DJ, Gage F. Stem and progenitor cells: [24] Wang HS, Hung SC, Peng ST, et al. Mesenchymal stem cells in the
origins, phenotypes, lineage commitments, and transdifferentiati- Wharton's jelly of the human umbilical cord. Stem Cells 2004; 22:
ons. Annu Rev Cell Dev Biol 2001; 17: 387-403. 1330-7.
[2] Kondo M, Wagers AJ, Manz MG, et al. Biology of hematopoietic [25] Weiss ML, Medicetty S, Bledsoe AR, et al. Human umbilical cord
stem cells and progenitors: implications for clinical application. matrix stem cells: preliminary characterization and effect of
Annu Rev Immunol 2003; 21: 759-806. transplantation in a rodent model of Parkinson's disease. Stem Cells
[3] Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, 2006; 24: 781-92.
Keiliss-Borok IV. Stromal cells responsible for transferring the [26] Xu J, Liao W, Gu D, Liang L, et al. Neural ganglioside GD2
microenvironment of the hemopoietic tissues. Cloning in vitro and identifies a subpopulation of mesenchymal stem cells in umbilical
retransplantation in vivo. Transplantation 1974; 12: 331-40. cord. Cell Physiol Biochem 2009; 23: 415-24.
[4] Prockop DJ. Marrow stromal cells as stem cells for [27] Karahuseyinoglu S, Cinar O, Kilic E, et al. Biology of stem cells in
nonhematopoietic tissues. Science 1997; 276: 71-4. human umbilical cord stroma: in situ and in vitro surveys. Stem
[5] Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential Cells 2007; 25: 319-31.
of adult human mesenchymal stem cells. Science 1999; 284: 143-7. [28] Baksh D, Yao R, Tuan RS. Comparison of proliferative and
[6] Delorme B, Ringe J, Pontikoglou C, et al. Specific lineage-priming multilineage differentiation potential of human mesenchymal stem
of bone marrow mesenchymal stem cells provides the molecular cells derived from umbilical cord and bone marrow. Stem Cells
framework for their plasticity. Stem Cells 2009; 12: 1142-51. 2007; 25: 1384-92.
[7] Sacchetti B, Funari A, Michienzi S, et al. Self-renewing [29] Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE.
osteoprogenitors in bone marrow sinusoids can organize a Human umbilical cord perivascular (HUCPV) cells: a source of
hematopoietic microenvironment. Cell 2007; 131: 324-36. mesenchymal progenitors. Stem Cells 2005; 23: 220-9.
[8] Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for [30] Moodley Y, Atienza D, Manuelpillai U, et al. Human umbilical
defining multipotent mesenchymal stromal cells. The International cord mesenchymal stem cells reduce fibrosis of bleomycin-induced
Society for cellular therapy position statement. Cytotherapy 2006; lung injury. Am J Pathol 2009; 175: 303-13.
8: 315-7. [31] Pereira WC, Khushnooma I, Madkaikar M, Ghosh K. Reproducible
[9] Caplan AI. Mesenchymal stem cells. J Orthop Res 1991; 9(5): 641- methodology for the isolation of mesenchymal stem cells from
50. human umbilical cord and its potential for cardiomyocyte
[10] da Silva Meirelles L Chagastelles PC, Nardi NB. Mesenchymal generation. J Tissue Eng Regen Med 2008; 2: 394-9.
stem cells reside in virtually all post-natal organs and tissues. J Cell [32] Mitchell KE, Weiss ML, Mitchell BM, et al. Matrix cells from
Sci 2006; 119: 2204-13. Wharton's jelly form neurons and glia. Stem Cells 2003; 21: 50-60.
[11] Anzalone R, Corrao S, Lo Iacono M, et al. Isolation and [33] Majore I, Moretti P, Stahl F, Hass R, Kasper C. Growth and
Characterization of CD276+/HLA-E+ Human Subendocardial differentiation properties of mesenchymal stromal cell populations
Mesenchymal Stem Cells from Chronic Heart Failure Patients: derived from whole human umbilical cord. Stem Cell Rev 2011; 7:
Analysis of Differentiative Potential and Immunomodulatory 17-31.
Markers Expression. Stem Cells Dev 2013; 22: 1-17. [34] La Rocca G, Anzalone R, Corrao S, et al. Isolation and
[12] Phinney DG, Prockop DJ. Concise review: mesenchymal characterization of Oct-4+/HLA-G+ mesenchymal stem cells from
stem/multipotent stromal cells: the state of transdifferentiation and human umbilical cord matrix: differentiation potential and
modes of tissue repair--current views. Stem Cells 2007; 25: 2896- detection of new markers. Histochem Cell Biol 2009; 131: 267-82.
902. [35] De Bruyn C, Najar M, Raicevic G, et al. A rapid, simple, and
[13] Bianco P, Robey PG, Saggio I, Riminucci M. "Mesenchymal" stem reproducible method for the isolation of mesenchymal stromal cells
cells in human bone marrow (skeletal stem cells): a critical from Wharton's jelly without enzymatic treatment. Stem Cells Dev
discussion of their nature, identity, and significance in incurable 2011; 20: 547-57.
skeletal disease. Hum Gene Ther 2010; 21: 1057-66. [36] Salehinejad P, Alitheen NB, Ali AM, et al. Comparison of different
[14] Castro-Malaspina H, Gay RE, Resnick G, et al. Characterization of methods for the isolation of mesenchymal stem cells from human
human bone marrow fibroblast colony-forming cells (CFU-F) and umbilical cord Wharton's jelly. In vitro Cell Dev Biol Anim 2012;
their progeny. Blood 1980; 56: 289-301. 48: 75-83.
[15] Mueller SM, Glowacki J. Age-related decline in the osteogenic [37] Carlin R, Davis D, Weiss M, Schultz B, Troyer D. Expression of
potential of human bone marrow cells cultured in three- early transcription factors Oct-4, Sox-2 and Nanog by porcine
dimensional collagen sponges. J Cell Biochem 2001; 82: 583-90. umbilical cord (PUC) matrix cells. Reprod Biol Endocrinol 2006;
[16] Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B, 4: 8.
Hows JM. Adult bone marrow is a rich source of human [38] Hoynowski SM, Fry MM, Gardner BM, et al. Characterization and
mesenchymal stem cells but umbilical cord and mobilized blood differentiation of equine umbilical cord-derived matrix cells.
adult blood are not. Br J Haematol 2003; 121: 368-74. Biochem Biophys Res Commun 2007; 362: 347-53.
[17] Perdikogianni C, Dimitriou H, Stiakaki E, Martimianaki G, [39] Nekanti U, Rao VB, Bahirvani AG, Jan M, Totey S, Ta M. Long-
Kalmanti M. Could cord blood be a source of mesenchymal term expansion and pluripotent marker array analysis of Wharton's
stromal cells for clinical use? Cytotherapy 2008; 10: 452-9. jelly-derived mesenchymal stem cells. Stem Cells Dev 2010; 19:
[18] Troyer DL, Weiss ML. Concise review: Wharton's jelly-derived 117-30.
cells are a primitive stromal cell population. Stem Cells 2008; 26: [40] Zhang ZY, Teoh SH, Chong MS, et al. Superior osteogenic
591-9. capacity for bone tissue engineering of fetal compared with
[19] Nanaev AK, Kohnen G, Milovanov AP, Domogatsky SP, perinatal and adult mesenchymal stem cells. Stem Cells 2009; 27:
Kaufmann P. Stromal differentiation and architecture of the human 126-37.
umbilical cord. Placenta 1997; 18: 53-64. [41] Hsieh JY, Fu YS, Chang SJ, Tsuang YH, Wang HW. Functional
[20] Wang XY, Lan Y, He WY, et al. Identification of mesenchymal module analysis reveals differential osteogenic and stemness
stem cells in aorta-gonad-mesonephros and yolk sac of human potentials in human mesenchymal stem cells from bone marrow
embryos. Blood 2008; 111: 2436-43. and Wharton's jelly of umbilical cord. Stem Cells Dev 2010; 19:
[21] Can A, Karahuseyinoglu S. Concise review: human umbilical cord 1895-910.
Stroma with Regard to the source of fetus-derived stem cells. Stem [42] Huang YC, Parolini O, La RG, Deng L. Umbilical cord versus
Cells 2007; 25: 2886-95. bone marrow-derived mesenchymal stromal cells. Stem Cells Dev
[22] Fong CY, Richards M, Manasi N, Biswas A, Bongso A. 2012; 21: 2900-3.
Comparative growth behaviour and characterization of stem cells
154 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.

[43] Chen MY, Lie PC, Li ZL, Wei X. Endothelial differentiation of [62] Yoo KH, Jang IK, Lee MW, et al. Comparison of
Wharton's jelly-derived mesenchymal stem cells in comparison immunomodulatory properties of mesenchymal stem cells derived
with bone marrow-derived mesenchymal stem cells. Exp Hematol from adult human tissues. Cell Immunol 2009; 259: 150-6.
2009; 37: 629-40. [63] Najar M, Raicevic G, Boufker HI, et al. Mesenchymal stromal cells
[44] Lo Iacono M, Anzalone R, Corrao S, et al. Perinatal and Wharton's use PGE2 to modulate activation and proliferation of lymphocyte
Jelly-Derived Mesenchymal Stem Cells in Cartilage Regenerative subsets: Combined comparison of adipose tissue, Wharton's Jelly
Medicine and Tissue Engineering Strategies. Open Tissue Eng and bone marrow sources. Cell Immunol 2010; 264: 171-9.
Regen Med J 2011; 4: 72-81. [64] Chen K, Wang D, Du WT, et al. Human umbilical cord
[45] Wang L, Tran I, Seshareddy K, Weiss ML, Detamore MS. A mesenchymal stem cells hUC-MSCs exert immunosuppressive
comparison of human bone marrow-derived mesenchymal stem activities through a PGE2-dependent mechanism. Clin Immunol
cells and human umbilical cord-derived mesenchymal stromal cells 2010; 135: 448-58.
for cartilage tissue engineering. Tissue Eng Part A 2009; 15: 2259- [65] Pontikoglou C, Deschaseaux F, Sensebe L, Papadaki HA. Bone
66. Marrow Mesenchymal Stem Cells: Biological Properties and Their
[46] Wu KH, Zhou B, Lu SH, et al. In vitro and in vivo differentiation Role in Hematopoiesis and Hematopoietic Stem Cell
of human umbilical cord derived stem cells into endothelial cells. J Transplantation. Stem Cell Rev 2011; 7: 569-89.
Cell Biochem 2007; 100: 608-16. [66] Valencic E, Piscianz E, Andolina M, Ventura A, Tommasini A.
[47] Conconi MT, Burra P, Di LR, et al. CD105(+) cells from The immunosuppressive effect of Wharton's jelly stromal cells
Wharton's jelly show in vitro and in vivo myogenic differentiative depends on the timing of their licensing and on lymphocyte
potential. Int J Mol Med 2006; 18: 1089-96. activation. Cytotherapy 2010; 12: 154-60.
[48] Zhao Z, Chen Z, Zhao X, et al. Sphingosine-1-phosphate promotes [67] Che N, Li X, Zhou S, et al. Umbilical cord mesenchymal stem cells
the differentiation of human umbilical cord mesenchymal stem suppress B-cell proliferation and differentiation. Cell Immunol
cells into cardiomyocytes under the designated culturing 2012; 274: 46-53.
conditions. J Biomed Sci 2011; 18: 37. [68] Ennis J, Gotherstrom C, Le Blanc K, Davies JE. In vitro
[49] Martin-Rendon E, Sweeney D, Lu F, Girdlestone J, Navarrete C, immunologic properties of human umbilical cord perivascular cells.
Watt SM. 5-Azacytidine-treated human mesenchymal Cytotherapy 2008; 10: 174-81.
stem/progenitor cells derived from umbilical cord, cord blood and [69] Gluckman E, Broxmeyer HA, Auerbach AD, et al. Hematopoietic
bone marrow do not generate cardiomyocytes in vitro at high reconstitution in a patient with Fanconi's anemia by means of
frequencies. Vox Sang 2008; 95: 137-48. umbilical-cord blood from an HLA-identical sibling. N Engl J Med
[50] Qian Q, Qian H, Zhang X, et al. 5-Azacytidine induces cardiac 1989; 321: 1174-8.
differentiation of human umbilical cord-derived mesenchymal stem [70] Gluckman E. Current status of umbilical cord blood hematopoietic
cells by activating extracellular regulated kinase. Stem Cells Dev stem cell transplantation. Exp Hematol 2000; 28: 1197-205.
2012; 21: 67-75. [71] Mayani H, Lansdorp PM. Biology of human umbilical cord blood-
[51] Ma L, Feng XY, Cui BL, et al. Human umbilical cord Wharton's derived hematopoietic stem/progenitor cells. Stem Cells 1998; 16:
Jelly-derived mesenchymal stem cells differentiation into nervelike 153-65.
cells. Chin Med J 2005; 118: 1987-93. [72] Migliaccio AR, Adamson JW, Stevens CE, Dobrila NL, Carrier
[52] Fu YS, Cheng YC, Lin MY, et al. Conversion of human umbilical CM, Rubinstein P. Cell dose and speed of engraftment in
cord mesenchymal stem cells in Wharton's jelly to dopaminergic placental/umbilical cord blood transplantation: graft progenitor cell
neurons in vitro: potential therapeutic application for Parkinsonism. content is a better predictor than nucleated cell quantity. Blood
Stem Cells 2006; 24: 115-24. 2000; 96: 2717-22.
[53] Anzalone R, Lo Iacono M, Corrao S, et al. New emerging [73] Gluckman E, Rocha V, Arcese W, et al. Factors associated with
potentials for human Wharton's jelly mesenchymal stem cells: outcomes of unrelated cord blood transplant: guidelines for donor
immunological features and hepatocyte-like differentiative choice. Exp Hematol 2004; 32: 397-407.
capacity. Stem Cells Dev 2010; 19: 423-38. [74] Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of
[54] Campard D, Lysy PA, Najimi M, Sokal EM. Native umbilical cord cord-blood transplantation from related and unrelated donors.
matrix stem cells express hepatic markers and differentiate into Eurocord Transplant Group and the European Blood and Marrow
hepatocyte-like cells. Gastroenterology 2008; 134: 833-48. Transplantation Group. N Engl J Med 1997; 337: 373-81.
[55] Wu LF, Wang NN, Liu YS, Wei X. Differentiation of Wharton's [75] Almeida-Porada G, Flake AW, Glimp HA, Zanjani ED.
jelly primitive stromal cells into insulin-producing cells in Cotransplantation of stroma results in enhancement of engraftment
comparison with bone marrow mesenchymal stem cells.Tissue Eng and early expression of donor hematopoietic stem cells in utero.
Part A 2009; 15: 2865-73. Exp Hematol 1999; 27: 1569-75.
[56] Chao KC, Chao KF, Fu YS, Liu SH. Islet-like clusters derived [76] Tung SS, Parmar S, Robinson SN, De LM, Shpall EJ. Ex vivo
from mesenchymal stem cells in Wharton's Jelly of the human expansion of umbilical cord blood for transplantation. Best Pract
umbilical cord for transplantation to control type 1 diabetes. PLoS Res Clin Haematol 2010;23:245-57.
One 2008; 3: e1451. [77] McNiece I, Harrington J, Turney J, Kellner J, Shpall EJ. Ex vivo
[57] Anzalone R, Lo Iacono M, Loria T, et al. Wharton's Jelly expansion of cord blood mononuclear cells on mesenchymal stem
Mesenchymal Stem Cells as Candidates for Beta Cells cells. Cytotherapy 2004; 6: 311-7.
Regeneration: Extending the Differentiative and [78] Robinson SN, Ng J, Niu T, et al. Superior ex vivo cord blood
Immunomodulatory Benefits of Adult Mesenchymal Stem Cells for expansion following co-culture with bone marrow-derived
the Treatment of Type 1 Diabetes. Stem Cell Reviews and Reports mesenchymal stem cells. Bone Marrow Transplant 2006; 37: 359-
2011; 7: 342-63. 66.
[58] La Rocca G. Connecting the Dots: The Promises of Wharton's Jelly [79] Walenda T, Bokermann G, Ventura Ferreira MS, et al. Synergistic
Mesenchymal Stem Cells for Tissue Repair and Regeneration. The effects of growth factors and mesenchymal stromal cells for
Open Tissue Engineering and Regenerative Medicine Journal 2011; expansion of hematopoietic stem and progenitor cells. Exp
12: 3-5. Hematol 2011; 39: 617-28.
[59] Deuse T, Stubbendorff M, Tang-Quan K, et al. Immunogenicity [80] Robinson SN, Simmons PJ, Yang H, Alousi AM, de Lima JM,
and immunomodulatory properties of umbilical cord lining Shpall EJ. Mesenchymal stem cells in ex vivo cord blood
mesenchymal stem cells. Cell Transplant 2011; 20: 655-67. expansion. Best Pract Res Clin Haematol 2011; 24: 83-92.
[60] Zhou C, Yang B, Tian Y, et al. Immunomodulatory effect of [81] Friedman R, Betancur M, Boissel L, Tuncer H, Cetrulo C,
human umbilical cord Wharton's jelly-derived mesenchymal stem Klingemann H. Umbilical cord mesenchymal stem cells: adjuvants
cells on lymphocytes. Cell Immunol 2011; 272: 33-8. for human cell transplantation. Biol Blood Marrow Transplant
[61] Weiss ML, Anderson C, Medicetty S, et al. Immune properties of 2007; 13: 1477-86.
human umbilical cord Wharton's jelly-derived cells. Stem Cells [82] Bakshi T, Zabriskie RC, Bodie S, et al. Mesenchymal stem cells
2008; 26: 2865-74. from Whartin's jelly of umbilical cord segments provide stromal
support for the maintenance of cord blood hematopoietic stem cells
during long-term ex vivo culture. Transfusion 2008; 48: 2638-44.
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 155

[83] Fong CY, Gauthaman K, Cheyyatraivendran S, Lin HD, Biswas A, with potential for clinical and tissue engineering applications. Adv
Bongso A. Human umbilical cord Wharton's jelly stem cells and its Biochem Eng Biotechnol 2010; 123: 29-54.
conditioned medium support hematopoietic stem cell expansion ex [101] Mariotti E, Mirabelli P, Abate G, et al. Comparative characteristics
vivo. J Cell Biochem 2012; 113: 658-68. of mesenchymal stem cells from human bone marrow and placenta:
[84] Bacigalupo A. Management of acute graft-versus-host disease. Br J CD10, CD49d, and CD56 make a difference. Stem Cells Dev 2008;
Haematol 2007; 137: 87-98. 17: 1039-41.
[85] Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host [102] Delorme B, Ringe J, Gallay N, et al. Specific plasma membrane
disease. Lancet 2009; 373: 1550-61. protein phenotype of culture-amplified and native human bone
[86] Ringden O, Le Blanc K. Mesenchymal stem cells for treatment of marrow mesenchymal stem cells. Blood 2008; 111: 2631-5.
acute and chronic graft-versus-host disease, tissue toxicity and [103] Farias VA, Linares-Fernandez JL, Penalver JL, et al. Human
hemorrhages. Best Pract Res Clin Haematol 2011; 24: 65-72. umbilical cord stromal stem cell express CD10 and exert
[87] Wu KH, Chan CK, Tsai C, et al. Effective treatment of severe contractile properties. Placenta 2011; 32: 86-95
steroid-resistant acute graft-versus-host disease with umbilical [104] Ding DC, Shyu WC, Chiang MF, et al. Enhancement of
cord-derived mesenchymal stem cells. Transplantation neuroplasticity through upregulation of beta1-integrin in human
2011;91:1412-6. umbilical cord-derived stromal cell implanted stroke model.
[88] Choi SW, Levine JE, Ferrara JL. Pathogenesis and management of Neurobiol Dis 2007; 27: 339-53.
graft-versus-host disease. Immunol Allergy Clin North Am 2010; [105] Prasanna SJ, Gopalakrishnan D, Shankar SR, Vasandan AB.
30: 75-101. Proinflammatory cytokines, IFNgamma and TNFalpha, influence
[89] Kolb HJ. Graft-versus-leukemia effects of transplantation and immune properties of human bone marrow and Wharton jelly
donor lymphocytes. Blood 2008; 112: 4371-83. mesenchymal stem cells differentially. PLoS One 2010; 5: e9016.
[90] Sprangers B, Fevery S, Van WB, De Somer L, Waer M, Billiau [106] Qiao C, Xu W, Zhu W, et al. Human mesenchymal stem cells
AD. Can graft-versus-leukemia reactivity be dissociated from graft- isolated from the umbilical cord. Cell Biol Int 2008; 32: 8-15.
versus-host disease? Front Biosci 2007; 12: 4568-94. [107] Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE.
[91] Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor Human mesenchymal stem cells self-renew and differentiate
natural killer cell alloreactivity in mismatched hematopoietic according to a deterministic hierarchy. PLoS One 2009; 4: e6498.
transplants. Science 2002; 295: 2097-100. [108] Angelucci S,Marchisio M,Di Giuseppe F, et al. Proteome analysis
[92] Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell of human Wharton's jelly cells during in vitro expansion.
alloreactivity in HLA-mismatched hematopoietic stem cell ProteomeSci 2010; 8: 18.
transplantation. Blood 1999; 94(1): 333-9. [109] Gindraux F, Selmani Z, Obert L, et al. Human and rodent bone
[93] Boissel L, Tuncer HH, Betancur M, Wolfberg A, Klingemann H. marrow mesenchymal stem cells that express primitive stem cells
Umbilical cord mesenchymal stem cells increase expansion of cord mark primitive stem cell markers can be directly enriched by using
blood natural killer cells. Biol Blood Marrow Transplant 2008; 14: the CD49a molecule. Cell Tissue Res 2007; 327: 471-83.
1031-8. [110] Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O.
[94] Sun L, Akiyama K, Zhang H, et al. Mesenchymal stem cell HLA expression and immunologic properties of differentiated and
transplantation reverses multiorgan dysfunction in systemic lupus undifferentiated mesenchymal stem cells. Exp Hematol 2003; 3:
erythematosus mice and humans. Stem Cells 2009; 27: 1421-32. 890-6.
[95] Sun L, Wang D, Liang J, et al. Umbilical cord mesenchymal stem [111] Selmani Z, Naji A, Zidi I, et al. Human leukocyte antigen-G5
cell transplantation in severe and refractory systemic lupus secretion by human mesenchymal stem cells is required to suppress
erythematosus. Arthritis Rheum 2010; 62: 2467-75. T lymphocyte and natural killer function and to induce
[96] Shi D, Wang D, Li X, et al. Allogeneic transplantation of umbilical CD4+CD25highFOXP3+ regulatory T cells. Stem Cells 2008; 26:
cord-derived mesenchymal stem cells for diffuse alveolar 212-22.
hemorrhage in systemic lupus erythematosus. Clin Rheumatol [112] Struys T, Moreels M, Martens W, Donders R, Wolfs E,
2012; 31: 841-6. LambrichtsI. Ultrastructural and immunocytochemical analysis of
[97] Rachakatla RS, Pyle MM, Ayuzawa R, et al. Combination multilineag differentiated human dental pulp- and umbilical cord-
treatment of human umbilical cord matrix stem cell-based derived mesenchymal stem cells. Cells Tissues Organs 2011; 193:
interferon-beta gene therapy and 5-fluorouracil significantly 366-78.
reduces growth of metastatic human breast cancer in SCID mouse [113] Simmons PJ, Torok-Storb B. Identification of stromal cell
lungs. Cancer Invest 2008; 26: 662-70. precursors in human bone marrow by a novel monoclonal antibody,
[98] Ayuzawa R, Doi C, Rachakatla RS, et al. Naive human umbilical STRO-1. Blood 1991; 78: 55-62.
cord matrix derived stem cells significantly attenuate growth of [114] Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro
human breast cancer cells in vitro and in vivo. Cancer Lett 2009; RC. SSEA-4 identifies mesenchymal stem cells from bone marrow.
280: 31-7. Blood 2007; 109: 1743-51.
[99] Gauthaman K, Fong CY, Arularasu S, et al. Human Wharton's jelly [115] Riekstina U, Cakstina I, Parfejevs V, et al. Embryonic stem cell
stem cell conditioned medium and cell-free lysate inhibit human marker expression pattern in human mesenchymal stem cells
osteosarcoma and mammary carcinoma cell growth in vitro and in derived from bone marrow, adipose tissue, heart and dermis. Stem
xenograft mice. J Cell Biochem 2013; 114: 366-77. Cell Rev 2009; 5: 378-86.
[100] Moretti P, Hatlapatka T, Marten D, et al. Mesenchymal stromal
cells derived from human umbilical cord tissues: primitive cells

Received: October 16, 2012 Revised: December 17, 2012 Accepted: December 28, 2012

S-ar putea să vă placă și