SlideShare a Scribd company logo
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; 2
Graduate 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
Stem cells are defined by their unique capacity to self-
renew, differentiate into one or more cell lineages and tissue
types, and functionally repopulate a tissue in vivo [1]. On the
basis of their ability to give rise to one or more cell types,
known as potency, stem cells can be hierarchically classified
as unipotent, multipotent, pluripotent and totipotent. Unipo-
tent stem cells can produce only one cell type (e.g. neurons),
whereas multipotent ones can differentiate into several cell
types (e.g. hematopoietic stem cells, HSCs). An embryonic
stem cell (ESC), derived from the inner cell mass (ICM) of
the embryo is pluripotent as can differentiate into cells of the
three germ layers, but not into trophoblastic cells, whereas
the zygote is totipotent as it produces cells of embryonic as
well as extraembryonic tissue.
HSCs isolated from post-natal bone marrow (BM) and
umbilical cord blood (UCB) are generally acknowledged as
the prototype of multipotent adult tissue stem cells, being
able to differentiate into all mature blood lineages as well as
serially reconstitute hematopoiesis in lethally irradiated mice
at the single cell level [2]. In the late 60s and early 70s,
*Address correspondence to this author at the Department of Hematology,
University Hospital of Heraklion, P.O. Box 1352, Heraklion, Crete, Greece;
Tel: +30 2810 394629 ; Fax: +30 2810 394632 ;
E-mail: pontikoglou@yahoo.com
Friedenstein et al. demonstrated that a population of non-
hematopoietic, plastic-adherent, fibroblast-like cells can also
be isolated from BM [3]. These cells, which were later
shown to be able to give rise to adipocytes, chondrocytes,
osteocytes, smooth muscle cells, fibroblasts and hema-
topoietic supportive stroma [3-6] and be self-renewing [7],
are currently referred to as mesenchymal stem/stromal cells
(MSCs) [4, 5, 8, 9]. So far MSC-like cells have been isolated
from several tissues, such as skeletal muscle, adipose tissue,
umbilical cord, synovium, dental pulp, amniotic fluid, as
well as fetal blood, liver, BM, lung and heart [10-12]. Never-
theless, these populations are not equivalent in terms of mul-
tipotency, when in vivo assays are used [13].
The field of MSCs has witnessed considerable progress
over the last decade. Due to the unraveling of many of their
exceptional characteristics and the encouraging preclinical
and clinical studies, it is now widely accepted that MSCs
will provide a revolutionary advantage in the therapeutic
intervention of various diseases or tissue damage in the near
future. More than forty years since their initial description,
BM-MSCs represent the most extensively studied population
of adult MSCs and are still considered as the gold-standard
for MSC clinical applications. Nevertheless it is now becom-
ing increasingly clear that BM may not represent the most
suitable source for MSC collection. Besides the invasive and
painful isolation procedure, MSCs are present at very low
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
and quantitative properties decline with donor's age [15].
The low frequency implies that an extensive in vitro expan-
sion of cells is usually required to obtain sufficient cell num-
bers for therapeutic interventions, thereby enhancing the risk
of epigenetic damages as well as viral and bacterial contami-
nations. To circumvent these obstacles, much effort has been
invested over the last decade in identifying alternative, more
abundant and easily attainable sources of MSCs for thera-
peutic use. In this regard the umbilical cord (UC), which is
considered medical waste and is obtained with a painless,
simple and safe procedure during delivery, has aroused much
attention during the last years as a promising source for MSC
isolation. MSCs have actually been found in both UC blood
(UCB) as well as in UC tissues. Nevertheless, the scarcity of
MSC in UCB (their frequency being much lower than in
BM) and the lack of a robust and reproducible technique for
harvesting and expanding MSCs from UCB units [16, 17]
have hampered their use in clinical applications. On the other
hand existing isolation protocols have constantly induced the
harvest of large numbers of MSCs from UC-tissues with a
hundred per cent success.
UC-MSCs share many properties with adult BM-MSCs,
but are generally considered as a more primitive population
than the latter. In fact, while they possess several characteris-
tics of ESCs, they do not form teratomas upon transplanta-
tion [18] and their research does not raise ethical or legal
issues. All these features open attractive perspectives for the
propagation and engineering of UC-derived MSCs for cell-
based therapies. This review will first attempt to give a
summary of the biology of MSCs derived from Wharton's
Jelly (WJ-MSCs), the most abundant region of UC and then
discuss their potential application for the treatment of cancer
and immune mediated disorders such as graft versus host
disease (GVHD) and systemic lupus erythematosus (SLE).
Finally, their putative role as feeder layers for ex vivo hema-
topoietic stem cell (HSC) expansion will be pointed out.
Human Umbilical Cord is a Rich Source of MSCs
The human UC (Fig. 1) contains two arteries and a vein
buried within a mucous or gelatinous connective tissue also
known as the Wharton’s jelly (WJ), enclosed by a simple
amniotic epithelium. WJ is largely made from proteoglycans
and various types of collagen, forming a sponge-like tissue,
within which stromal cells are embedded [19]. These cells,
also referred to as umbilical cord matrix (UCM) cells are
thought to be trapped into the WJ early in embryogenesis
during their migration from the aortic-gonadotropin-
mesonephric region to the fetal liver through the umbilical
cord [20]. UCM cells have exceptional properties in that
although they are bona fide MSCs [18, 21] as discussed be-
low in detail, possessing similar properties with their adult
BM counterparts, they also retain characteristics of primitive
stem cells, like the expression of embryonic stem cell mark-
ers [22, 23]. In line with these observations MSCs derived
from WJ, hereafter referred to as "UC-MSCs", maintain their
self-renewal capacity in vitro for a longer period of time, and
exhibit a shorter doubling time and a broader pluripotency as
compared to BM-MSCs [18]. These unique developmental
and operational characteristics along with the fact that UC-
MSCs can be easily isolated, extracorporeally without risks
for the donor, from a tissue which is routinely discarded,
highlight the advantages of UC-MSCs over ESCs and adult
MSCs and explain the rapidly growing interest in this field
of stem cell biology.
The first report providing robust evidence that WJ stro-
mal cells can be classified as MSCs was published in 2004
[24]. In that study, cells extracted from WJ, displayed a fi-
broblast-like phenotype when expanded in vitro and were
able to differentiate into osteoblasts, adipocytes and chon-
drocytes upon appropriate stimulation [24]. Furthermore,
akin to BM-MSCs they were shown to express high levels of
CD29, CD44, CD51, CD73 and CD105, but lacked expres-
sion of the hematopoietic markers CD34 and CD45 (Table 1)
[24]. Subsequent reports have provided additional informa-
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.
A
Wha
A
U bili l iUmbilical arteries
Perivascular
Intervasc
Amniotic epithelium
arton’s Jelly
Umbilical vein/
Umbilical cord blood
Amniotic epithelium
Subam
cular
nion
146 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al.
tion on WJ stromal cell phenotype, demonstrating the ex-
pression of various membrane molecules known to be asso-
ciated with a MSC-like identity. These molecules include
CD90, CD146, CD166 and ganglioside 2 (GD2) [8, 23, 25,
26]. Collectively, the morphology, immunophenotype and
differentiation potential of WJ stromal cells fulfill the criteria
for MSCs proposed by the International Society for Cellular
Therapy (ISCT) [8]. Indeed, given that at present there is no
specific marker to define UC-MSCs, the expression of the
aforementioned set of membrane antigens combined with the
demonstration of in vitro multi-lineage differentiation poten-
tial is necessary to identify MSCs in UC-derived tissues
UC-MSCs have been isolated from three relatively indis-
tinct regions of the WJ: the perivascular, the intervascular
and the subamnion zones (Fig. 1) [27]. Cells from these
zones share a fibroblast-like morphology, a high proliferative
rate and multipotent differentiation capacity. However a
comprehensive head-to-head comparison between MSCs
obtained from different regions of the UC has not been per-
formed thus far, due to the great variation in cell isola-
tion/culture protocols. Hence the question whether cells har-
vested from different components of the UC represent differ-
ent populations is still open. Nevertheless one study combin-
ing both in situ and in vitro experiments [27] has demon-
strated that MSCs derived from the perivascular region com-
prise a distinct subpopulation than those derived from the
intervascular and subamniotic parts of the UC. It has thus
been reported that perivascular MSCs stain strongly for pan-
cytokeratin as compared to intervascular and subamniotic
counterparts and give rise in vitro to flat, wide cytoplasmic
cells, named type 1 cells, which tend to diminish in number
during protracted culture [27]. On the other hand intervascu-
lar and subamniotic MSCs generate fibroblast-like cells per-
sisting throughout culture (type 2 cells). In view of these
findings it appears that UC-MSCs are actually type 2 cells,
displaying adipogenic, chondrogenic, osteogenic and neu-
ronal capacity, upon appropriate differentiation stimuli [27].
Isolation of MSCs from UC perivascular zone has been re-
ported by other groups as well [28, 29]. These cells, termed
as Human Umbilical Cord Perivascular Cells (HUCPVCs),
have a colony forming unit (CFU) frequency of 1:333 [29]
and differ in their growth characteristics from UC-MSCs
(population doubling times ranging from 59.4±42.4h in pas-
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
population of UC-MSCs [27, 29]. Furthermore, HUCPVCs
exhibit higher proliferative capacity and osteogenic potential
as compared to BM-MSCs [28]. The phenotype of
HUCPVCs shares many similarities with that of WJ-derived
MSCs (Table 1). One significant difference though, is the
high expression of CD146 by HUCPVCs. Interestingly this
marker is either absent [28] or weakly expressed by MSCs
derived form other regions of WJ [30].
Isolation of MSCs from Wharton's Jelly
In recent years, several methods (Fig. 2) have been de-
veloped for the isolation of MSCs from WJ. However, ex-
perimental approaches vary considerably and in some cases
harvested cells from UC tissue most likely consist of a het-
erogeneous population, thereby biasing comparisons. With
regards to protocols used to isolate UC-MSCs, some authors
remove umbilical cord vessels, scrape off WJ with a scalpel
and then treat the tissue enzymatically [24]. Others cut down
UC to smaller segments, strip the vessels and then directly
immerse the remaining UC to an enzymatic solution [25, 27,
31]. Interestingly, neither the enzymes used nor the incuba-
tion period have been standardized thus far. Alternatively,
instead of enzymatic digestion, several groups dissect the
cord segment into very small pieces, with or without discard-
ing the cord vessels and the segments are used as explants
from which cultures are subsequently established [32-34].
The major advantage of this method is that it minimizes cell
damage as it does not involve enzymatic treatment, however
it should be noted that primary cultures generated in this way
are quite heterogeneous [33]. UC-MSCs have also been iso-
lated from WJ by simply cutting UC segments longitudinally
and plating them with WJ side-down onto a plastic surface,
once again without enzyme digestion or dissection [35]. Fi-
nally, to collect HUCPVCs some groups remove blood ves-
sels together with surrounding WJ, tie up the ends of each
vessel into a loop and then immerse the vessel in enzyme,
thereby isolating cells from the perivascular tissue [28, 29].
The question as to which is the optimal method for effi-
cient isolation of a homogeneous and well defined popula-
tion of UC-MSCs has not been answered thus far. However,
unless an agreement is reached between laboratories regard-
ing the isolation and ex vivo expansion of UC-MSCs, there
will be no consensus on the characteristics and biological
properties of the cells, which is a prerequisite for the proper
design of preclinical studies, so as to pave the way for the
potential use of UC-MSCs in clinical applications.
Do differences in isolation procedures and ex vivo expan-
sion protocols have an impact on the antigens expressed by
UC-MSCs? This issue has not been properly investigated so
far, yet a recent study [36] demonstrated that MSC markers
CD73, CD90 and CD105 varied their expression patterns
according to whether enzyme cocktails or explants were used
for UC-MSC harvesting. A similar variation with respect to
the isolation method was also noted in the expression of the
ESC markers c-Kit and Oct-4 by UC-MSCs [36].
WJ-derived UC-MSCs may be Regarded as a Primitive
Stromal Cell Population
Porcine UC-MSCs have been reported to express the
ESC markers Oct-4, Sox-2 and Nanog [37] and equine UC-
MSCs express, apart from Oct-4, the embryonic markers
SSEA4 and c-Kit [38]. In line with these findings, human
UC-MSCs express Oct-4, Nanog, Sox-2, c-Kit, as well as
Dnmt3b and Tert, albeit at lower levels as compared to ESCs
[22, 34, 39]. Such a low expression of the aforementioned
pluripotency markers may explain why UC-MSCs do not
produce teratomas in vivo in immunodeficient mice and rats
[22, 25]. In addition human UC-MSCs express the ESC
markers Tra-1-60, Tra-1-81, SSEA-1 and SSEA-4 [22].
Many of these pluripotent stem cell markers retain their ex-
pression for at least nine passages during ex vivo expansion.
Comparative gene expression profiling between UC-MSCs
and BM-MSCs demonstrated that the former express higher
levels of Nanog, Dnmt3b and Gabrb3, and also
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]
CD54 ICAM-1 (intercellular adhesion molecule-1)
- [47]
+ [105]
+ [29] + [5]
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]
CD106 VCAM-1 (Vascular cell adhesion molecule-1)
- [49]
+ [61]
- [29]
+ [107]
+ [5]
CD117 C-kit receptor
- [104]
+ [108]
- [49]
+ [29]
- [102]
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]
STRO-1
- [81]
+ [112]
+ [113]
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]
Fig. (2). A scheme depicting the different approaches followed for the establishment of WJ-derived UC-MSC cultures (modified from [100]).
Umbilical cord
Cut tissue into
very small pieces
Dissection in
small segments
Explant culture
Stromal cell culture
Filter
Removal of umbilical
vessels
Scrapping off
mesenchymal tissue
Enzymatic treatment
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
markers Brix, CD9, Gal, Kit and Rex1 [39]. The implication
of these markers in the biology of UC-MSCs is currently
unknown, yet their expression is thought to reflect the more
primitive nature of UC-MSCs as compared to their adult
counterparts within the BM. However, not all UC-derived
MSCs express ESC markers. In fact, it has been demon-
strated that primitive markers are expressed only by a subset
of UC-MSCs [26, 40], thereby suggesting that these cells are
rather hetereogeneous in terms of stemness. It would there-
fore be very challenging to identify that specific marker
which would permit the isolation of the primi-
tive/multipotent MSCs from UC.
Human UC-MSCs exhibit also higher expression of
genes implicated in the phosphoinositide 3-kinase (PI3K)-
AKT survival/proliferation pathway, including GAB2,
NFKBIA, FOXO1A, YWHAB, FOXO3A, CDKN1A,
MAP3K8, NFKB1, NOS3, DDR1 and TEK [41]. Further-
more, the NF -B pathway has been reported to be more ac-
tive in UC-MSCs than in BM-MSCs [41]. These findings
together with the fact that human WJ-MSCs express the
primitive stem cell marker TERT [25] explain, at least in
part the observation that UC-MSCs have a shorter doubling
time [27] and a more extensive ex vivo expansion capacity
[18].
Multilineage in vitro Differentiation Potential of WJ-
derived UC-MSCs
Accumulating evidence suggests that UC-MSCs can be
successfully induced to differentiate into mesodermal and
non-mesodermal lineages. As mentioned above, several
groups have established that UC-MSCs can generate os-
teoblasts, adipocytes and chondrocytes in vitro [24, 27, 42,
43], thereby fulfilling one of the minimal criteria proposed
by ISCT for MSCs [8]. However, lipid accumulation takes
much longer in UC-MSCs and the adipocytes are less mature
than those derived from BM-MSCs [27, 41]. In addition,
UC-MSCs have been shown to possess weaker osteogenic
[41] and chondrogenic [44, 45] potential as compared to
BM-MSCs.
UC-MSCs have also been reported to successfully differ-
entiate into endothelial cells [43, 46] and actually demon-
strate higher endothelial differentiation potential when com-
pared to BM-MSCs [43]. In accordance with this observation
non-differentiated UC-MSCs exhibit higher expression of
endothelial associated genes including FLT1, GATA4,
GATA6, ISL1, LAMA1, SOX17 and SERPINA1 [39].
One group has suggested, that UC-MSCs can give rise to
skeletal muscle cells when cultured in the appropriate me-
dium and that non-differentiated UC-MSCs may display
myogenic potential in vivo [47]. Differentiation of UC-MSCs
to cardiomyocytes has also been reported but is still contro-
versial [24, 31, 48-50]. For instance, one group [24] demon-
strated that UC-MSCs could be appropriately induced to
form cells demonstrating cardiomyocyte morphology and
expressing cardiac markers, including N-cadherin and car-
diac troponin. Another team [49], however, was unable to
detect cardiomyocyte marker expression after in vitro induc-
tion of MSCs isolated from WJ and perivascular tissues. De-
finitive evidence that UC-MSC-derived cardiomyocytes rep-
resent functional and terminally differentiated cells is still
scarce. To our knowledge, only one group has thus far re-
ported that UC-MSC-derived cardiomyocytes exhibit slight
spontaneous beating after 21 days of induction [31].
Finally, several groups have shown that UC-MSCs can
acquire morphological and biochemical properties of neural
cells when exposed to appropriate differentiation media [27,
32, 51, 52]. Mitchell et al. [32] were the first who observed
UC-MSC neuronal differentiation, following a complex and
multi-step neuronal induction process. Differentiation was
assessed by changes in morphology and expression of neu-
ron specific enolase, bIII-tubulin, neurofilament and tyrosin
hydroxylase. Subsequent studies have further corroborated
these findings. It has also been suggested that UC-MSCs
may be more primed for neuronal differentiation as com-
pared to BM-MSCs, since they express, higher levels of neu-
ronal-associated genes including NESTIN, GFAP and
SEM3A [39] in a non–differentiated state. However, it
should be kept in mind that definitive evidence for electro-
physiological activity of UC-MSC-derived neurons is still
missing. Therefore the issue whether the neuronal-like phe-
notype of UC-MSCs reflects a genuine trans-differentiation
is still pending.
Recent studies suggest that UC-MSCs can also differen-
tiate into endodermal lineages (reviewed in [53]). In this
context, Campard et al. [54] reported that non-differentiated
UCM cells constitutively expressed markers of hepatic line-
age, such as albumin, alpha-fetoprotein, cytokeratin-19, con-
nexin-32, and dipeptidyl peptidase IV. After in vitro hepatic
induction, cells exhibited hepatocyte-like morphology and
upregulated the expression of certain hepatocyte-associated
markers, but lacked important functional properties of ma-
ture liver cells, thereby questioning whether UCM cells have
actually undergone hepatocyte maturation [54]. Other groups
have also claimed that UC-MSCs can be induced to form
isle-like cells [55, 56] (reviewed in [57]), able to express
pancreatic associated genes and release insulin in response to
physiological glucose levels [56]. Interestingly, UC-MSCs
have been reported to have a higher pancreatic differentia-
tion potential than their BM counterparts [55].
The Immunomodulatory Properties of UC-MSCs
Apart from the multilineage potential, UC-MSCs possess
various immunoregulatory properties, rendering them as
appealing candidates for cell based therapies [58]. More pre-
cisely UC-MSCs have been found to exhibit very low ex-
pression of HLA class I, and lack the expression of HLA-DR
[59, 60]. Notably, in comparison to BM-MSCs derived from
aged donors, UC-MSCs express significantly lower levels of
HLA-I, and exhibit significantly higher intracellular concen-
trations of the immunosuppressive molecule HLA-G [59].
These findings together with the observations that UC-MSCs
do not express the co-stimulatory molecules CD40, CD80
and CD86 [59, s61], and do not elicit proliferative responses
from allogeneic T cells in vitro [61], suggest that UC-MSCs
may be characterized by inherently low immunogenicity.
However, another study suggests that UC-MSCs may not be
as immune-privileged as thought, as they can trigger weak
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].
Similar findings have been reported for BM-MSCs as well.
Interestingly the latter have been shown to be much more
immunocompetent compared to UC-MSCs [59].
Several studies have been published on the immunosup-
pressive potential of UC-MSCs on T lymphocytes [59, 60,
62-64]. One group reported that UC-MSCs target CD4+ and
CD8+ T subpopulations equally and are able to inhibit T-cell
proliferation regardless of the stimulus used to activate T-
cells [63]. Furthermore, UC-MSC-mediated inhibitory ef-
fects were higher than with BM-MSCs. In accordance with
previous studies suggesting that the immunomodulatory ef-
fect of MSCs on T cells may be mediated, at least in part, by
soluble factors (reviewed in [65]), such as hepatocyte growth
factor (HGF), prostaglandin E2 (PGE2), transforming growth
factor- 1 (TGF- 1) and indoleamine 2,3-dioxygenase (IDO),
the authors assessed whether any of these molecules may be
implicated in UC-MSC immunoregulation as well. In their
hands immune suppression exerted by both UC- and BM-
MSCs was mediated by cycloxygenase (COX) 1 and COX2
enzymes, regulating PGE2 synthesis, and by the production
of PGE2 per se, whereas HGF did not seem to be actively
implicated. These findings were further corroborated by an-
other study [64] which additionally showed that while UC-
MSCs intrinsically secrete very low levels of PGE2, its pro-
duction is upregulated when cells are co-cultured in the pres-
ence of activated human peripheral blood mononuclear cells
(hPBMCs). The authors attributed the PGE2 stimulation to
inflammatory cytokines (including IFN- and interleukin-1 )
produced by hPBMCs [64].
Interestingly, several studies [59, 62, 66] have reported
that UC-MSC immune characteristics may be modified fol-
lowing exposure to inflammatory cytokines and/or activated
PBMCs/T-cells so as to display immunosupressive proper-
ties, a process described by the term licensing or priming. In
this context, Deuse et al. [59] reported that IFN- at doses
below 50ng/ml upregulated HLA-DR and HLA-I expression,
thereby increasing UC-MSC immunogenicity. IFN- treat-
ment also induced tolerogenicity by increasing intracellular
HLA-G expression, TGF- and IL-10 release, and stimulated
the production of IDO [59]. Higher doses of IFN- however
enhanced UC-MSC immunosuppressive phenotype by
downregulating HLA-DR expression and further increasing
IDO. These results suggest that appropriate priming may be
crucial for the fine adjustment of MSC characteristics, ren-
dering them suitable for a given therapeutic application.
The immunomodulatory effects of MSCs are not only
apparent in T lymphocytes but also concern B lymphocytes.
To our knowledge however, the impact of UC-MSCs on B
cells has been poorly investigated and undoubtedly awaits
further elucidation, as the information that will be gathered is
expected to provide crucial clues for the introduction of WJ-
MSCs into the clinic. In the single study reported until today
[67] UC-MSCs were found to inhibit B cell proliferation,
differentiation and antibody production in vitro. This sup-
pressive effect is mediated, at least in part by soluble factors,
which may downregulate Blimp-1 and upregualate PAX-5,
two master regulators of B cell differentiation [67]. Further-
more UC-MSCs have been demonstrated to inhibit Akt and
p38 MAPK phosphorylation. Interestingly, these signal
transduction pathways are implicated in the regulation of B
cell proliferation and differentiation [67].
The immunoregulatory potential of HUCPVCs has also
been assessed in a recent study [68]. More precisely it has
been demonstrated that when HUCPVCs are co-cultured
with resting or stimulated allogeneic lymphocytes they fail to
induce their proliferation. Furthermore, HUCPVCs have
been found to reduce in vitro alloreactivity in mixed lym-
phocyte reactions. Notably, the immunosuppressive effect of
the cells is exerted, at least in part via soluble factors [68].
UC-MSCs as Feeder Layers for ex vivo Umbilical Cord
Blood Expansion
Despite the fact that BM has been the most extensively
studied source for HSC transplantation, the importance of
UCB as an alternative source of transplantable HSCs has
been widely acknowledged since the first successful UCB
transplant in 1988 [69]. From a practical point of view UCB
transplantation has several advantages over BM, including
the non-invasiveness of the procedure, the ease of procure-
ment, the reduced risk of transmissible infections and the
availability for immediate use [70]. In addition, UCB grafts
allow for a greater tolerable HLA disparity with a decreased
frequency of acute or chronic graft versus host disease
(GVHD) as compared to BM grafts [70]. Finally UCB con-
tains a higher proportion of immature and committed hema-
topoietic progenitor cells (HPCs) in comparison to adult BM
[71].
However UCB transplantation has also some significant
restrictions, limiting its broad applicability. The most sig-
nificant limiting factor of UCB use is probably the low total
cell dose harvested from a UCB unit, which in turn is associ-
ated with delayed neutrophil and platelet engraftment and
higher engraftment failure [72]. For this reason UCB trans-
plantation remains significantly more successful in children
[73], whereas poor outcomes have been reported in patients
>45 kgr who receive only one unit of UCB [74].
To circumvent these obstacles, so as to improve out-
comes and extend the application of UCB transplantation to
older and/or larger patients, several groups have explored the
possibility of in vivo expansion of HSCs from UCB units.
Various protocols are under investigation over the last 30
years, including those based on cultures on feeder layers.
Among the different cell types that have been used as stro-
mal support for UCB expansion, the most extensively stud-
ied thus far are BM- MSCs.
The rationale behind the use of MSCs as feeder cells for
UCB expansion lies on a large body of evidence, clearly
demonstrating the BM-MSC promoting role on hematopoie-
sis (reviewed in [65]). More precisely BM-MSCs have been
shown in vivo to enhance HSC engraftment and repopula-
tion, as evidenced by the observation that co-transplantation
of human HSCs and MSCs results in increased chimerism
and/or hematopoietic recovery, in both animal models and
humans [65, 75]. On the other hand, MSCs have been shown
in vitro to maintain long-term culture initiating cells (LT-
CICs) and expand lineage-specific colony forming units
from CD34+
marrow cells in long-term BM cultures (re-
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
been elucidated, it is now widely accepted that MSCs supply
HSCs and their progeny with signals for survival, prolifera-
tion and differentiation through direct cell-to-cell contact
and/or production of cytokines, adhesion molecules and ex-
tracellular matrix proteins (reviewed in [65]).
These observations have provided the theoretical back-
ground for the investigation of the potential use of MSCs as
scaffolds for the ex vivo expansion of HSCs. Based on the
hypothesis that MSCs could provide many of the molecular
cues of the hematopoietic microenvironment necessary to
favor HSC self-renewal and regulate their proliferation, CB
hematopoetic stem/progenitor cells have been co-cultured on
BM-MSC preformed layers in a medium containing (FBS)
and various combinations of growth factors including G-
CSF, stem cell factor (SCF), thrombopoietin (TPO), Flt3-
ligand or fibroblast growth factor-1 (FGF-1) [76]. Using this
ex vivo model several groups have demonstrated an increase
in the number of total nucleated cells (TNC), clonogenic
cells or CD34+
cells [77, 78]. These findings were further
corroborated in a recent study [79], where CB CD34+
cells
were co-cultured with BM-MSCs in a serum free medium
supplemented with SCF, TPO and FGF-1. BM-MSCs were
also shown to maintain the primitive phenotype of CB hema-
topoietic stem and progenitor cells even after massive expan-
sion [79]. However co-culture with BM-MSCs did not result
in a significant expansion of long-term repopulating cells. A
clinical trial is underway at M.D. Anderson Cancer Center
combining an unmanipulated UCB unit with an expanded
UCB unit for 14 days in the presence of a cytokine cocktail
on layer of family member donor-derived BM-MSCs or "off-
the-self" BM-MSCs (Angioblast Systems Inc.). Preliminary
results suggest a beneficial role for these stromal cell-based
HSC expansion [80].
Given the promising role of BM-MSCs in supporting ex
vivo UCB expansion and since UC-MSCs share many char-
acteristics with their BM counterparts, but in contrast to the
latter can be readily and painlessly harvested from UC, they
soon emerged as attractive candidates for HSC expansion. In
this regard, the first report demonstrating that akin to BM-
MSCs, UC-MSCs can also support hematopoiesis was pub-
lished in 2007 [81]. In that study the authors showed that
UC-MSCs secrete significantly higher amounts of several
important cytokines and hematopoietic growth factors in-
cluding G-CSF, GM-CSF, LIF, IL-1 , L-6, IL-8 and IL-11,
compared to BM-MSCs. Furthemore co-culture of UC-
MSCs with UCB CD34+
cells significantly increased hema-
topoietic colony formation as compared to clonogenic pro-
genitor cell assays performed in the absence of MSCs [81].
Finally, in agreement with previous reports demonstrating
that co-transplantation of BM-MSCs with HSCs can acceler-
ate hematopoietic engraftment, co-injection of UC-MSCs
with UCB cells or CD34+
cells in a NOD/SCID mouse
model accelerated human HSC recovery. The authors as-
sumed that the improved engraftment might be attributed to
the presence of UC- MSCs within the BM of the recipient
animals and also to the production of hematopoietic cytoki-
nes by these cells. Subsequent reports have confirmed the
capacity of UC-MSCs and UC-MSC conditioned medium to
support the growth and maintenance of UCB hematopoietic
stem/progenitor cells in long term culture [82, 83], thereby
establishing an ex vivo model that could enable the study of
the interactions between HSCs and their supportive micro-
environment within the UC. However, whether UC MSC-
based HSC expansion will have possible future clinical util-
ity is currently unknown and awaits further clarification.
UC-MSCs for GVHD Treatment
Graft versus host disease (GVHD) is one of the most
severe complications of allogeneic BM transplantation. First-
line treatment with corticosteroids is the gold standard for
this T-cell mediated process, however second line treatment
for steroid-resistant GVHD has not been clearly established
thus far and the outcome is poor, with one year overall sur-
vival (OS) of only 30% [84, 85].
The in vitro and in vivo immunomodulatory properties of
BM-MSCs have provided the theoretical background for the
use of these cells for treatment of acute steroid-resistant
GVHD. Several reports have been published thus far (re-
viewed in [86]) suggesting that clinical responses can actu-
ally be attained in a significant number of patients, especially
in children. BM-MSCs have also been used for the treatment
of chronic GVHD showing likewise promising results [86].
To our knowledge, there has been only one recent study
clinically testing UC- MSCs for GVHD [87]. In that particu-
lar report third-party culture expanded UC- MSCs were
transplanted in 2 children with severe steroid-resistant acute
GVHD. The first patient received three infusions of UC-
MSCs. Each time acute GVHD improved significantly and
eventually disappeared. The second patient received only
one infusion of UC-MSCs that led to complete remission of
acute GVHD. No child experienced any adverse events what
so ever and both children were well and remained off immu-
nosuppressive therapy at the time the study was published.
The authors concluded that UC-MSCs may be considered as
a feasible, safe and effective treatment modality for acute
GVHD [87]. These encouraging results need obviously to be
confirmed by larger trials, which will include adult patients
as well. Furthermore, additional studies are needed in order
to optimize the use of UC-MSCs in treating GVHD. For ex-
ample, it is currently unknown which is the optimal UC-
MSC dose and schedule, or whether the use of platelet-
lysate, or other substitutes of the standard FBS-enriched me-
dium (so as to comply with regulatory guidelines for the
production of MSCs for clinical use, which recommend
avoiding the use of animal derivatives) might influence the
therapeutic effect of UC-MSCs.
UC-MSCs Support the Expansion of Natural Killer Cells
As previously mentioned a major toxicity associated with
Bone Marrow Transplantation (BMT) is GVHD, an attack
by the T cells in the graft against the recipient organs includ-
ing the skin, gastrointestinal tract, liver, and eyes [88]. Ag-
gressive depletion of T cells in the allogeneic graft can
markedly reduce the incidence of acute GVHD, however,
this is associated with significant drawbacks, including re-
duction of the graft versus tumor effect of the allogeneic-
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-
come [90]. Given that natural killer (NK) cells have been
found to control leukemia relapse while protecting patients
from GVHD [91, 92], the possibility of expanding and po-
tentially using these cells for adoptive immunotherapy ap-
pears an attractive approach in the setting of allogeneic
BMT. Provided that UCB is rich in NK cell progenitors and
since UC-MSCs can produce significant amounts of cytoki-
nes, Boissel et al. [93] hypothesized that a feeder layer of
UC-MSCs might actually effectively support ex vivo expan-
sion of UCB-NK cells. Indeed when CD3-depleted UCB
mononuclear cells were cocultured with UC-MSCs and a
cytokine cocktail, the proportion of CD3-
CD56+
NK cells
increased significantly as compared to cell cultured with
cytokines only. Interestingly direct contact between UCB
mononuclear cells and UC-MSCs resulted in a significantly
greater NK cell expansion. Notably, expanded NK cells
maintained an elevated cytotoxic profile [93]. The possibility
of further optimizing this experimental protocol should be
evaluated in additional studies and of course the cytotoxicity
of the expanded NK cells awaits assessment in animal mod-
els.
UC-MSC Transplantation in Systemic Lupus Erythema-
tosus
To our knowledge, apart from GVHD, in vivo therapeutic
efficacy of UC- MSC transplantation has also been clearly
illustrated in the setting of systemic lupus erythematosus
(SLE). The theoretical background for assessing the clinical
impact of UC-MSCs in patients with this autoimmune disor-
der has been provided by the immunomodulatory properties
of UC-MSCs and also by the beneficial effects of BM- MSC
administration in patients with treatment-refractory SLE
[94]. In this context, it has recently been reported that trans-
plantation of UC-MSCs in patients with severe and refrac-
tory SLE results in clinical and serologic improvement and is
not associated with mortality or other treatment-related ad-
verse events [95]. Even though the mechanisms mediating
the therapeutic effect of UC-MSCs in SLE patients remain
unclear, it has been suggested that disease remission may be
associated with an increase in the proportion of Tregs in pe-
ripheral blood along with a decrease in IL-4 serum levels,
probably resulting in inhibition of humoral activity [95].
Another recent study has demonstrated that UC-MSCs can
also provide clinical benefit in diffuse alveolar hemorrhage,
a rare complication of SLE with high clinical mortality [96].
These findings suggest that UC-MSC transplantation may
hold promise as a treatment modality in selected SLE cases.
However, the efficacy and safety of this strategy need to be
further validated in larger clinical trials with longer periods
of follow-up.
Therapeutic Potential of UC-MSCs in Cancer
An emerging clinical application of UC-MSCs that re-
cently gained much attention is their use as cellular therapy
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
severe combined immunodeficiency (SCID) mice, bearing
human breast carcinoma tumors in their lungs [97]. Inter-
estingly, UC-MSCs were only found in the lungs and not in
other tissues. This combined approach resulted in significant
metastatic tumor reduction, much greater than compared to
each treatment alone. A more recent study has corroborated
the ability of UC-MSCs to reduce the growth of human
breast carcinoma cells both in vitro as well as in vivo, in a
mouse xenograft model [98]. Notably, the antitumor proper-
ties of UC-MSCs do not seem to be specific only to breast
cancer cells, but have also been demonstrated in osteosar-
coma and ovarian carcinoma cell lines in vitro [99]. The
mechanisms mediating UC-MSC anticancer effects are still
under investigation. However, it has been suggested that UC-
MSCs exert their antitumor activity both via cell-to-cell con-
tact and humoral factors, through induction of apoptosis, cell
cycle arrest and autophagy [97, 99]. Taken together, the
aforementioned preliminary data imply that UC-MSCs may
hold promise as a therapeutic modality in cancer and high-
light the need for the development of additional well-
designed pre-clinical studies so as to properly investigate the
potential of this cell-based approach.
CONCLUSION
Within recent years considerable interest has been given
to the exciting properties of UC-MSCs, derived from the WJ,
because of their primitive nature as compared to BM-MSCs
(the prototype of adult MSCs), their multilineage potential
across germ layers, the extensive in vitro proliferation and
their immunosuppressive capacity. These characteristics
along with the ease by which UC-MSCs are isolated from a
tissue with unlimited availability, have generated much en-
thusiasm regarding their potential applications in cell-based
therapies. This expectation is being supported by both pre-
and clinical data. In particular, in this review we reported on
the promising results obtained in recent studies regarding the
use of UC-MSCs for the treatment of cancer and immune
mediated disorders, such as graft versus host disease and
systemic lupus erythematosus and discussed UC-MSC role
as feeder layers for ex vivo HSC expansion. Nevertheless
much work has to be done so as to fully unravel the biologic
features of UC-MSCs and especially those distinguishing
them from adult MSCs. Furthermore, properly designed
comparative studies need to be undertaken in the near future
so as to clearly identify and characterize MSC subpopula-
tions originating from different regions of WJ, in order to
answer the long-standing question: whether different ana-
tomical locations within WJ are associated with distinct
qualitative and quantitative MSC properties. Notably, avail-
able global expression technologies, like gene expression
and comparative genomic hybridization (CGH) microarrays,
proteome and secretome profiles, methylation mapping and
ChIP-on-ChiP technologies are anticipated to contribute sig-
nificantly to the better understanding of UC-MSC biology. It
is anticipated that the data obtained by these studies will not
only provide the theoretical background for the eventual in-
troduction of UC-MSCs in the clinic, but would also be ma-
jor contribution to stem cell biology in general.
CONFLICT OF INTEREST
The authors confirm that this article content has no con-
flicts of interest.
Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 153
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Weissman IL, Anderson DJ, Gage F. Stem and progenitor cells:
origins, phenotypes, lineage commitments, and transdifferentiati-
ons. Annu Rev Cell Dev Biol 2001; 17: 387-403.
[2] Kondo M, Wagers AJ, Manz MG, et al. Biology of hematopoietic
stem cells and progenitors: implications for clinical application.
Annu Rev Immunol 2003; 21: 759-806.
[3] Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF,
Keiliss-Borok IV. Stromal cells responsible for transferring the
microenvironment of the hemopoietic tissues. Cloning in vitro and
retransplantation in vivo. Transplantation 1974; 12: 331-40.
[4] Prockop DJ. Marrow stromal cells as stem cells for
nonhematopoietic tissues. Science 1997; 276: 71-4.
[5] Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential
of adult human mesenchymal stem cells. Science 1999; 284: 143-7.
[6] Delorme B, Ringe J, Pontikoglou C, et al. Specific lineage-priming
of bone marrow mesenchymal stem cells provides the molecular
framework for their plasticity. Stem Cells 2009; 12: 1142-51.
[7] Sacchetti B, Funari A, Michienzi S, et al. Self-renewing
osteoprogenitors in bone marrow sinusoids can organize a
hematopoietic microenvironment. Cell 2007; 131: 324-36.
[8] Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for
defining multipotent mesenchymal stromal cells. The International
Society for cellular therapy position statement. Cytotherapy 2006;
8: 315-7.
[9] Caplan AI. Mesenchymal stem cells. J Orthop Res 1991; 9(5): 641-
50.
[10] da Silva Meirelles L Chagastelles PC, Nardi NB. Mesenchymal
stem cells reside in virtually all post-natal organs and tissues. J Cell
Sci 2006; 119: 2204-13.
[11] Anzalone R, Corrao S, Lo Iacono M, et al. Isolation and
Characterization of CD276+/HLA-E+ Human Subendocardial
Mesenchymal Stem Cells from Chronic Heart Failure Patients:
Analysis of Differentiative Potential and Immunomodulatory
Markers Expression. Stem Cells Dev 2013; 22: 1-17.
[12] Phinney DG, Prockop DJ. Concise review: mesenchymal
stem/multipotent stromal cells: the state of transdifferentiation and
modes of tissue repair--current views. Stem Cells 2007; 25: 2896-
902.
[13] Bianco P, Robey PG, Saggio I, Riminucci M. "Mesenchymal" stem
cells in human bone marrow (skeletal stem cells): a critical
discussion of their nature, identity, and significance in incurable
skeletal disease. Hum Gene Ther 2010; 21: 1057-66.
[14] Castro-Malaspina H, Gay RE, Resnick G, et al. Characterization of
human bone marrow fibroblast colony-forming cells (CFU-F) and
their progeny. Blood 1980; 56: 289-301.
[15] Mueller SM, Glowacki J. Age-related decline in the osteogenic
potential of human bone marrow cells cultured in three-
dimensional collagen sponges. J Cell Biochem 2001; 82: 583-90.
[16] Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B,
Hows JM. Adult bone marrow is a rich source of human
mesenchymal stem cells but umbilical cord and mobilized blood
adult blood are not. Br J Haematol 2003; 121: 368-74.
[17] Perdikogianni C, Dimitriou H, Stiakaki E, Martimianaki G,
Kalmanti M. Could cord blood be a source of mesenchymal
stromal cells for clinical use? Cytotherapy 2008; 10: 452-9.
[18] Troyer DL, Weiss ML. Concise review: Wharton's jelly-derived
cells are a primitive stromal cell population. Stem Cells 2008; 26:
591-9.
[19] Nanaev AK, Kohnen G, Milovanov AP, Domogatsky SP,
Kaufmann P. Stromal differentiation and architecture of the human
umbilical cord. Placenta 1997; 18: 53-64.
[20] Wang XY, Lan Y, He WY, et al. Identification of mesenchymal
stem cells in aorta-gonad-mesonephros and yolk sac of human
embryos. Blood 2008; 111: 2436-43.
[21] Can A, Karahuseyinoglu S. Concise review: human umbilical cord
Stroma with Regard to the source of fetus-derived stem cells. Stem
Cells 2007; 25: 2886-95.
[22] Fong CY, Richards M, Manasi N, Biswas A, Bongso A.
Comparative growth behaviour and characterization of stem cells
from human Wharton's jelly. Reprod Biomed Online 2007; 15:
708-18.
[23] Fong CY, Chak LL, Biswas A,et al. Human Wharton's jelly stem
cells have unique transcriptome profiles compared to human
embryonic stem cells and other mesenchymal stem cells. Stem Cell
Rev 20011; 7: 1-16.
[24] Wang HS, Hung SC, Peng ST, et al. Mesenchymal stem cells in the
Wharton's jelly of the human umbilical cord. Stem Cells 2004; 22:
1330-7.
[25] Weiss ML, Medicetty S, Bledsoe AR, et al. Human umbilical cord
matrix stem cells: preliminary characterization and effect of
transplantation in a rodent model of Parkinson's disease. Stem Cells
2006; 24: 781-92.
[26] Xu J, Liao W, Gu D, Liang L, et al. Neural ganglioside GD2
identifies a subpopulation of mesenchymal stem cells in umbilical
cord. Cell Physiol Biochem 2009; 23: 415-24.
[27] Karahuseyinoglu S, Cinar O, Kilic E, et al. Biology of stem cells in
human umbilical cord stroma: in situ and in vitro surveys. Stem
Cells 2007; 25: 319-31.
[28] Baksh D, Yao R, Tuan RS. Comparison of proliferative and
multilineage differentiation potential of human mesenchymal stem
cells derived from umbilical cord and bone marrow. Stem Cells
2007; 25: 1384-92.
[29] Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE.
Human umbilical cord perivascular (HUCPV) cells: a source of
mesenchymal progenitors. Stem Cells 2005; 23: 220-9.
[30] Moodley Y, Atienza D, Manuelpillai U, et al. Human umbilical
cord mesenchymal stem cells reduce fibrosis of bleomycin-induced
lung injury. Am J Pathol 2009; 175: 303-13.
[31] Pereira WC, Khushnooma I, Madkaikar M, Ghosh K. Reproducible
methodology for the isolation of mesenchymal stem cells from
human umbilical cord and its potential for cardiomyocyte
generation. J Tissue Eng Regen Med 2008; 2: 394-9.
[32] Mitchell KE, Weiss ML, Mitchell BM, et al. Matrix cells from
Wharton's jelly form neurons and glia. Stem Cells 2003; 21: 50-60.
[33] Majore I, Moretti P, Stahl F, Hass R, Kasper C. Growth and
differentiation properties of mesenchymal stromal cell populations
derived from whole human umbilical cord. Stem Cell Rev 2011; 7:
17-31.
[34] La Rocca G, Anzalone R, Corrao S, et al. Isolation and
characterization of Oct-4+/HLA-G+ mesenchymal stem cells from
human umbilical cord matrix: differentiation potential and
detection of new markers. Histochem Cell Biol 2009; 131: 267-82.
[35] De Bruyn C, Najar M, Raicevic G, et al. A rapid, simple, and
reproducible method for the isolation of mesenchymal stromal cells
from Wharton's jelly without enzymatic treatment. Stem Cells Dev
2011; 20: 547-57.
[36] Salehinejad P, Alitheen NB, Ali AM, et al. Comparison of different
methods for the isolation of mesenchymal stem cells from human
umbilical cord Wharton's jelly. In vitro Cell Dev Biol Anim 2012;
48: 75-83.
[37] Carlin R, Davis D, Weiss M, Schultz B, Troyer D. Expression of
early transcription factors Oct-4, Sox-2 and Nanog by porcine
umbilical cord (PUC) matrix cells. Reprod Biol Endocrinol 2006;
4: 8.
[38] Hoynowski SM, Fry MM, Gardner BM, et al. Characterization and
differentiation of equine umbilical cord-derived matrix cells.
Biochem Biophys Res Commun 2007; 362: 347-53.
[39] Nekanti U, Rao VB, Bahirvani AG, Jan M, Totey S, Ta M. Long-
term expansion and pluripotent marker array analysis of Wharton's
jelly-derived mesenchymal stem cells. Stem Cells Dev 2010; 19:
117-30.
[40] Zhang ZY, Teoh SH, Chong MS, et al. Superior osteogenic
capacity for bone tissue engineering of fetal compared with
perinatal and adult mesenchymal stem cells. Stem Cells 2009; 27:
126-37.
[41] Hsieh JY, Fu YS, Chang SJ, Tsuang YH, Wang HW. Functional
module analysis reveals differential osteogenic and stemness
potentials in human mesenchymal stem cells from bone marrow
and Wharton's jelly of umbilical cord. Stem Cells Dev 2010; 19:
1895-910.
[42] Huang YC, Parolini O, La RG, Deng L. Umbilical cord versus
bone marrow-derived mesenchymal stromal cells. Stem Cells Dev
2012; 21: 2900-3.
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
Wharton's jelly-derived mesenchymal stem cells in comparison
with bone marrow-derived mesenchymal stem cells. Exp Hematol
2009; 37: 629-40.
[44] Lo Iacono M, Anzalone R, Corrao S, et al. Perinatal and Wharton's
Jelly-Derived Mesenchymal Stem Cells in Cartilage Regenerative
Medicine and Tissue Engineering Strategies. Open Tissue Eng
Regen Med J 2011; 4: 72-81.
[45] Wang L, Tran I, Seshareddy K, Weiss ML, Detamore MS. A
comparison of human bone marrow-derived mesenchymal stem
cells and human umbilical cord-derived mesenchymal stromal cells
for cartilage tissue engineering. Tissue Eng Part A 2009; 15: 2259-
66.
[46] Wu KH, Zhou B, Lu SH, et al. In vitro and in vivo differentiation
of human umbilical cord derived stem cells into endothelial cells. J
Cell Biochem 2007; 100: 608-16.
[47] Conconi MT, Burra P, Di LR, et al. CD105(+) cells from
Wharton's jelly show in vitro and in vivo myogenic differentiative
potential. Int J Mol Med 2006; 18: 1089-96.
[48] Zhao Z, Chen Z, Zhao X, et al. Sphingosine-1-phosphate promotes
the differentiation of human umbilical cord mesenchymal stem
cells into cardiomyocytes under the designated culturing
conditions. J Biomed Sci 2011; 18: 37.
[49] Martin-Rendon E, Sweeney D, Lu F, Girdlestone J, Navarrete C,
Watt SM. 5-Azacytidine-treated human mesenchymal
stem/progenitor cells derived from umbilical cord, cord blood and
bone marrow do not generate cardiomyocytes in vitro at high
frequencies. Vox Sang 2008; 95: 137-48.
[50] Qian Q, Qian H, Zhang X, et al. 5-Azacytidine induces cardiac
differentiation of human umbilical cord-derived mesenchymal stem
cells by activating extracellular regulated kinase. Stem Cells Dev
2012; 21: 67-75.
[51] Ma L, Feng XY, Cui BL, et al. Human umbilical cord Wharton's
Jelly-derived mesenchymal stem cells differentiation into nervelike
cells. Chin Med J 2005; 118: 1987-93.
[52] Fu YS, Cheng YC, Lin MY, et al. Conversion of human umbilical
cord mesenchymal stem cells in Wharton's jelly to dopaminergic
neurons in vitro: potential therapeutic application for Parkinsonism.
Stem Cells 2006; 24: 115-24.
[53] Anzalone R, Lo Iacono M, Corrao S, et al. New emerging
potentials for human Wharton's jelly mesenchymal stem cells:
immunological features and hepatocyte-like differentiative
capacity. Stem Cells Dev 2010; 19: 423-38.
[54] Campard D, Lysy PA, Najimi M, Sokal EM. Native umbilical cord
matrix stem cells express hepatic markers and differentiate into
hepatocyte-like cells. Gastroenterology 2008; 134: 833-48.
[55] Wu LF, Wang NN, Liu YS, Wei X. Differentiation of Wharton's
jelly primitive stromal cells into insulin-producing cells in
comparison with bone marrow mesenchymal stem cells.Tissue Eng
Part A 2009; 15: 2865-73.
[56] Chao KC, Chao KF, Fu YS, Liu SH. Islet-like clusters derived
from mesenchymal stem cells in Wharton's Jelly of the human
umbilical cord for transplantation to control type 1 diabetes. PLoS
One 2008; 3: e1451.
[57] Anzalone R, Lo Iacono M, Loria T, et al. Wharton's Jelly
Mesenchymal Stem Cells as Candidates for Beta Cells
Regeneration: Extending the Differentiative and
Immunomodulatory Benefits of Adult Mesenchymal Stem Cells for
the Treatment of Type 1 Diabetes. Stem Cell Reviews and Reports
2011; 7: 342-63.
[58] La Rocca G. Connecting the Dots: The Promises of Wharton's Jelly
Mesenchymal Stem Cells for Tissue Repair and Regeneration. The
Open Tissue Engineering and Regenerative Medicine Journal 2011;
12: 3-5.
[59] Deuse T, Stubbendorff M, Tang-Quan K, et al. Immunogenicity
and immunomodulatory properties of umbilical cord lining
mesenchymal stem cells. Cell Transplant 2011; 20: 655-67.
[60] Zhou C, Yang B, Tian Y, et al. Immunomodulatory effect of
human umbilical cord Wharton's jelly-derived mesenchymal stem
cells on lymphocytes. Cell Immunol 2011; 272: 33-8.
[61] Weiss ML, Anderson C, Medicetty S, et al. Immune properties of
human umbilical cord Wharton's jelly-derived cells. Stem Cells
2008; 26: 2865-74.
[62] Yoo KH, Jang IK, Lee MW, et al. Comparison of
immunomodulatory properties of mesenchymal stem cells derived
from adult human tissues. Cell Immunol 2009; 259: 150-6.
[63] Najar M, Raicevic G, Boufker HI, et al. Mesenchymal stromal cells
use PGE2 to modulate activation and proliferation of lymphocyte
subsets: Combined comparison of adipose tissue, Wharton's Jelly
and bone marrow sources. Cell Immunol 2010; 264: 171-9.
[64] Chen K, Wang D, Du WT, et al. Human umbilical cord
mesenchymal stem cells hUC-MSCs exert immunosuppressive
activities through a PGE2-dependent mechanism. Clin Immunol
2010; 135: 448-58.
[65] Pontikoglou C, Deschaseaux F, Sensebe L, Papadaki HA. Bone
Marrow Mesenchymal Stem Cells: Biological Properties and Their
Role in Hematopoiesis and Hematopoietic Stem Cell
Transplantation. Stem Cell Rev 2011; 7: 569-89.
[66] Valencic E, Piscianz E, Andolina M, Ventura A, Tommasini A.
The immunosuppressive effect of Wharton's jelly stromal cells
depends on the timing of their licensing and on lymphocyte
activation. Cytotherapy 2010; 12: 154-60.
[67] Che N, Li X, Zhou S, et al. Umbilical cord mesenchymal stem cells
suppress B-cell proliferation and differentiation. Cell Immunol
2012; 274: 46-53.
[68] Ennis J, Gotherstrom C, Le Blanc K, Davies JE. In vitro
immunologic properties of human umbilical cord perivascular cells.
Cytotherapy 2008; 10: 174-81.
[69] Gluckman E, Broxmeyer HA, Auerbach AD, et al. Hematopoietic
reconstitution in a patient with Fanconi's anemia by means of
umbilical-cord blood from an HLA-identical sibling. N Engl J Med
1989; 321: 1174-8.
[70] Gluckman E. Current status of umbilical cord blood hematopoietic
stem cell transplantation. Exp Hematol 2000; 28: 1197-205.
[71] Mayani H, Lansdorp PM. Biology of human umbilical cord blood-
derived hematopoietic stem/progenitor cells. Stem Cells 1998; 16:
153-65.
[72] Migliaccio AR, Adamson JW, Stevens CE, Dobrila NL, Carrier
CM, Rubinstein P. Cell dose and speed of engraftment in
placental/umbilical cord blood transplantation: graft progenitor cell
content is a better predictor than nucleated cell quantity. Blood
2000; 96: 2717-22.
[73] Gluckman E, Rocha V, Arcese W, et al. Factors associated with
outcomes of unrelated cord blood transplant: guidelines for donor
choice. Exp Hematol 2004; 32: 397-407.
[74] Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of
cord-blood transplantation from related and unrelated donors.
Eurocord Transplant Group and the European Blood and Marrow
Transplantation Group. N Engl J Med 1997; 337: 373-81.
[75] Almeida-Porada G, Flake AW, Glimp HA, Zanjani ED.
Cotransplantation of stroma results in enhancement of engraftment
and early expression of donor hematopoietic stem cells in utero.
Exp Hematol 1999; 27: 1569-75.
[76] Tung SS, Parmar S, Robinson SN, De LM, Shpall EJ. Ex vivo
expansion of umbilical cord blood for transplantation. Best Pract
Res Clin Haematol 2010;23:245-57.
[77] McNiece I, Harrington J, Turney J, Kellner J, Shpall EJ. Ex vivo
expansion of cord blood mononuclear cells on mesenchymal stem
cells. Cytotherapy 2004; 6: 311-7.
[78] Robinson SN, Ng J, Niu T, et al. Superior ex vivo cord blood
expansion following co-culture with bone marrow-derived
mesenchymal stem cells. Bone Marrow Transplant 2006; 37: 359-
66.
[79] Walenda T, Bokermann G, Ventura Ferreira MS, et al. Synergistic
effects of growth factors and mesenchymal stromal cells for
expansion of hematopoietic stem and progenitor cells. Exp
Hematol 2011; 39: 617-28.
[80] Robinson SN, Simmons PJ, Yang H, Alousi AM, de Lima JM,
Shpall EJ. Mesenchymal stem cells in ex vivo cord blood
expansion. Best Pract Res Clin Haematol 2011; 24: 83-92.
[81] Friedman R, Betancur M, Boissel L, Tuncer H, Cetrulo C,
Klingemann H. Umbilical cord mesenchymal stem cells: adjuvants
for human cell transplantation. Biol Blood Marrow Transplant
2007; 13: 1477-86.
[82] Bakshi T, Zabriskie RC, Bodie S, et al. Mesenchymal stem cells
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,
Bongso A. Human umbilical cord Wharton's jelly stem cells and its
conditioned medium support hematopoietic stem cell expansion ex
vivo. J Cell Biochem 2012; 113: 658-68.
[84] Bacigalupo A. Management of acute graft-versus-host disease. Br J
Haematol 2007; 137: 87-98.
[85] Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host
disease. Lancet 2009; 373: 1550-61.
[86] Ringden O, Le Blanc K. Mesenchymal stem cells for treatment of
acute and chronic graft-versus-host disease, tissue toxicity and
hemorrhages. Best Pract Res Clin Haematol 2011; 24: 65-72.
[87] Wu KH, Chan CK, Tsai C, et al. Effective treatment of severe
steroid-resistant acute graft-versus-host disease with umbilical
cord-derived mesenchymal stem cells. Transplantation
2011;91:1412-6.
[88] Choi SW, Levine JE, Ferrara JL. Pathogenesis and management of
graft-versus-host disease. Immunol Allergy Clin North Am 2010;
30: 75-101.
[89] Kolb HJ. Graft-versus-leukemia effects of transplantation and
donor lymphocytes. Blood 2008; 112: 4371-83.
[90] Sprangers B, Fevery S, Van WB, De Somer L, Waer M, Billiau
AD. Can graft-versus-leukemia reactivity be dissociated from graft-
versus-host disease? Front Biosci 2007; 12: 4568-94.
[91] Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor
natural killer cell alloreactivity in mismatched hematopoietic
transplants. Science 2002; 295: 2097-100.
[92] Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell
alloreactivity in HLA-mismatched hematopoietic stem cell
transplantation. Blood 1999; 94(1): 333-9.
[93] Boissel L, Tuncer HH, Betancur M, Wolfberg A, Klingemann H.
Umbilical cord mesenchymal stem cells increase expansion of cord
blood natural killer cells. Biol Blood Marrow Transplant 2008; 14:
1031-8.
[94] Sun L, Akiyama K, Zhang H, et al. Mesenchymal stem cell
transplantation reverses multiorgan dysfunction in systemic lupus
erythematosus mice and humans. Stem Cells 2009; 27: 1421-32.
[95] Sun L, Wang D, Liang J, et al. Umbilical cord mesenchymal stem
cell transplantation in severe and refractory systemic lupus
erythematosus. Arthritis Rheum 2010; 62: 2467-75.
[96] Shi D, Wang D, Li X, et al. Allogeneic transplantation of umbilical
cord-derived mesenchymal stem cells for diffuse alveolar
hemorrhage in systemic lupus erythematosus. Clin Rheumatol
2012; 31: 841-6.
[97] Rachakatla RS, Pyle MM, Ayuzawa R, et al. Combination
treatment of human umbilical cord matrix stem cell-based
interferon-beta gene therapy and 5-fluorouracil significantly
reduces growth of metastatic human breast cancer in SCID mouse
lungs. Cancer Invest 2008; 26: 662-70.
[98] Ayuzawa R, Doi C, Rachakatla RS, et al. Naive human umbilical
cord matrix derived stem cells significantly attenuate growth of
human breast cancer cells in vitro and in vivo. Cancer Lett 2009;
280: 31-7.
[99] Gauthaman K, Fong CY, Arularasu S, et al. Human Wharton's jelly
stem cell conditioned medium and cell-free lysate inhibit human
osteosarcoma and mammary carcinoma cell growth in vitro and in
xenograft mice. J Cell Biochem 2013; 114: 366-77.
[100] Moretti P, Hatlapatka T, Marten D, et al. Mesenchymal stromal
cells derived from human umbilical cord tissues: primitive cells
with potential for clinical and tissue engineering applications. Adv
Biochem Eng Biotechnol 2010; 123: 29-54.
[101] Mariotti E, Mirabelli P, Abate G, et al. Comparative characteristics
of mesenchymal stem cells from human bone marrow and placenta:
CD10, CD49d, and CD56 make a difference. Stem Cells Dev 2008;
17: 1039-41.
[102] Delorme B, Ringe J, Gallay N, et al. Specific plasma membrane
protein phenotype of culture-amplified and native human bone
marrow mesenchymal stem cells. Blood 2008; 111: 2631-5.
[103] Farias VA, Linares-Fernandez JL, Penalver JL, et al. Human
umbilical cord stromal stem cell express CD10 and exert
contractile properties. Placenta 2011; 32: 86-95
[104] Ding DC, Shyu WC, Chiang MF, et al. Enhancement of
neuroplasticity through upregulation of beta1-integrin in human
umbilical cord-derived stromal cell implanted stroke model.
Neurobiol Dis 2007; 27: 339-53.
[105] Prasanna SJ, Gopalakrishnan D, Shankar SR, Vasandan AB.
Proinflammatory cytokines, IFNgamma and TNFalpha, influence
immune properties of human bone marrow and Wharton jelly
mesenchymal stem cells differentially. PLoS One 2010; 5: e9016.
[106] Qiao C, Xu W, Zhu W, et al. Human mesenchymal stem cells
isolated from the umbilical cord. Cell Biol Int 2008; 32: 8-15.
[107] Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE.
Human mesenchymal stem cells self-renew and differentiate
according to a deterministic hierarchy. PLoS One 2009; 4: e6498.
[108] Angelucci S,Marchisio M,Di Giuseppe F, et al. Proteome analysis
of human Wharton's jelly cells during in vitro expansion.
ProteomeSci 2010; 8: 18.
[109] Gindraux F, Selmani Z, Obert L, et al. Human and rodent bone
marrow mesenchymal stem cells that express primitive stem cells
mark primitive stem cell markers can be directly enriched by using
the CD49a molecule. Cell Tissue Res 2007; 327: 471-83.
[110] Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O.
HLA expression and immunologic properties of differentiated and
undifferentiated mesenchymal stem cells. Exp Hematol 2003; 3:
890-6.
[111] Selmani Z, Naji A, Zidi I, et al. Human leukocyte antigen-G5
secretion by human mesenchymal stem cells is required to suppress
T lymphocyte and natural killer function and to induce
CD4+CD25highFOXP3+ regulatory T cells. Stem Cells 2008; 26:
212-22.
[112] Struys T, Moreels M, Martens W, Donders R, Wolfs E,
LambrichtsI. Ultrastructural and immunocytochemical analysis of
multilineag differentiated human dental pulp- and umbilical cord-
derived mesenchymal stem cells. Cells Tissues Organs 2011; 193:
366-78.
[113] Simmons PJ, Torok-Storb B. Identification of stromal cell
precursors in human bone marrow by a novel monoclonal antibody,
STRO-1. Blood 1991; 78: 55-62.
[114] Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro
RC. SSEA-4 identifies mesenchymal stem cells from bone marrow.
Blood 2007; 109: 1743-51.
[115] Riekstina U, Cakstina I, Parfejevs V, et al. Embryonic stem cell
marker expression pattern in human mesenchymal stem cells
derived from bone marrow, adipose tissue, heart and dermis. Stem
Cell Rev 2009; 5: 378-86.
Received: October 16, 2012 Revised: December 17, 2012 Accepted: December 28, 2012

More Related Content

What's hot

Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)
ComprehensiveBiologi
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
ComprehensiveBiologi
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
ComprehensiveBiologi
 
Ms cand cryopreservation
Ms cand cryopreservationMs cand cryopreservation
Ms cand cryopreservation
ComprehensiveBiologi
 
Whartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_mWhartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_m
ComprehensiveBiologi
 
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Lipogems Equine & Lipogems Canine
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
ComprehensiveBiologi
 
Stem cells journal
Stem cells  journalStem cells  journal
Stem cells journal
Scidoc Publishers
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells research
Scidoc Publishers
 
Transplantation journal
Transplantation journalTransplantation journal
Transplantation journal
Scidoc Publishers
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterials
ComprehensiveBiologi
 
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
raditio ghifiardi
 
Cancer stem cells Cscs
Cancer stem cells CscsCancer stem cells Cscs
Cancer stem cells Cscs
Mahdy Ali Ahmad Osman
 
stem cells and cancer stem cells
 stem cells and cancer stem cells stem cells and cancer stem cells
stem cells and cancer stem cells
Marwa Khalifa
 
Breast Cancer Stem Cell - Basic
Breast Cancer Stem Cell - Basic Breast Cancer Stem Cell - Basic
Breast Cancer Stem Cell - Basic
Lobna Shash
 
Immunomodulatory properties of Mesenchymal Stem Cells
Immunomodulatory properties of Mesenchymal Stem CellsImmunomodulatory properties of Mesenchymal Stem Cells
Immunomodulatory properties of Mesenchymal Stem Cells
Shreya Ahuja
 
Stem cell research
Stem cell researchStem cell research
Stem cell research
Tariq Mohammed
 
Organoid culture in cancer
Organoid culture in cancerOrganoid culture in cancer
Organoid culture in cancer
Samieh Asadian
 
Trabalho 2012 teo2010 regenerative therapy
Trabalho 2012 teo2010 regenerative therapyTrabalho 2012 teo2010 regenerative therapy
Trabalho 2012 teo2010 regenerative therapy
Felemon Porto
 

What's hot (20)

Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
 
Ms cand cryopreservation
Ms cand cryopreservationMs cand cryopreservation
Ms cand cryopreservation
 
Whartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_mWhartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_m
 
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
 
Stem cells journal
Stem cells  journalStem cells  journal
Stem cells journal
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells research
 
Transplantation journal
Transplantation journalTransplantation journal
Transplantation journal
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterials
 
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
 
Cancer stem cells Cscs
Cancer stem cells CscsCancer stem cells Cscs
Cancer stem cells Cscs
 
stem cells and cancer stem cells
 stem cells and cancer stem cells stem cells and cancer stem cells
stem cells and cancer stem cells
 
Breast Cancer Stem Cell - Basic
Breast Cancer Stem Cell - Basic Breast Cancer Stem Cell - Basic
Breast Cancer Stem Cell - Basic
 
Application of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in MesenchymalApplication of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in Mesenchymal
 
Immunomodulatory properties of Mesenchymal Stem Cells
Immunomodulatory properties of Mesenchymal Stem CellsImmunomodulatory properties of Mesenchymal Stem Cells
Immunomodulatory properties of Mesenchymal Stem Cells
 
Stem cell research
Stem cell researchStem cell research
Stem cell research
 
Organoid culture in cancer
Organoid culture in cancerOrganoid culture in cancer
Organoid culture in cancer
 
Trabalho 2012 teo2010 regenerative therapy
Trabalho 2012 teo2010 regenerative therapyTrabalho 2012 teo2010 regenerative therapy
Trabalho 2012 teo2010 regenerative therapy
 

Similar to Whartons jelly review

International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
SciRes Literature LLC. | Open Access Journals
 
describe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdfdescribe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdf
feelingcomputors
 
A Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant CellsA Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant Cells
BRNSSPublicationHubI
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem Cells
DitoAnurogo1
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docx
joellemurphey
 
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson PublishersCancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
CrimsonpublishersCancer
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
gilpinleeanna
 
American Journal of Pharmacology & Therapeutics
American Journal of Pharmacology & TherapeuticsAmerican Journal of Pharmacology & Therapeutics
American Journal of Pharmacology & Therapeutics
SciRes Literature LLC. | Open Access Journals
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Lipogems Equine & Lipogems Canine
 
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiationSox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
Khalid Mukhtar
 
Mesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptxMesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptx
SwapnaliBehera
 
Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in Orthopaedics
Dr. Bushu Harna
 
tissue engineering ; basics and challenges.pdf
tissue engineering ; basics and challenges.pdftissue engineering ; basics and challenges.pdf
tissue engineering ; basics and challenges.pdf
Romissaa ali Esmail/ faculty of dentistry/Al-Azhar university
 
2. Isolation of stem cells and basic culture (1).pptx
2. Isolation of stem cells and basic culture (1).pptx2. Isolation of stem cells and basic culture (1).pptx
2. Isolation of stem cells and basic culture (1).pptx
PALLAVI895484
 
Cell Therapy: The New Approach to Dermatology and Dermatologic Surgery
Cell Therapy: The New Approach to Dermatology and Dermatologic SurgeryCell Therapy: The New Approach to Dermatology and Dermatologic Surgery
Cell Therapy: The New Approach to Dermatology and Dermatologic Surgery
semualkaira
 
Stem cells & therapy(mutaib)
Stem cells & therapy(mutaib)Stem cells & therapy(mutaib)
Stem cells & therapy(mutaib)
UNIVERSITY OF KASHMIR
 
Cancer diagnosis
Cancer diagnosisCancer diagnosis
Cancer diagnosis
Pavithra Balakrishnan
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
Ankita-rastogi
 

Similar to Whartons jelly review (20)

International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 
describe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdfdescribe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdf
 
A Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant CellsA Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant Cells
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem Cells
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docx
 
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson PublishersCancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
 
American Journal of Pharmacology & Therapeutics
American Journal of Pharmacology & TherapeuticsAmerican Journal of Pharmacology & Therapeutics
American Journal of Pharmacology & Therapeutics
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiationSox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
Sox2gene experssion on mouse embryonic stem cells in the neural diffrentiation
 
Mesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptxMesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptx
 
Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in Orthopaedics
 
tissue engineering ; basics and challenges.pdf
tissue engineering ; basics and challenges.pdftissue engineering ; basics and challenges.pdf
tissue engineering ; basics and challenges.pdf
 
2. Isolation of stem cells and basic culture (1).pptx
2. Isolation of stem cells and basic culture (1).pptx2. Isolation of stem cells and basic culture (1).pptx
2. Isolation of stem cells and basic culture (1).pptx
 
Cell Therapy: The New Approach to Dermatology and Dermatologic Surgery
Cell Therapy: The New Approach to Dermatology and Dermatologic SurgeryCell Therapy: The New Approach to Dermatology and Dermatologic Surgery
Cell Therapy: The New Approach to Dermatology and Dermatologic Surgery
 
Stem cells & therapy(mutaib)
Stem cells & therapy(mutaib)Stem cells & therapy(mutaib)
Stem cells & therapy(mutaib)
 
Cancer diagnosis
Cancer diagnosisCancer diagnosis
Cancer diagnosis
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
 

Recently uploaded

ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIONDACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
DR SETH JOTHAM
 
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness JourneyTom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
greendigital
 
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.GawadHemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
NephroTube - Dr.Gawad
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Savita Shen $i11
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
DR SETH JOTHAM
 
Novas diretrizes da OMS para os cuidados perinatais de mais qualidade
Novas diretrizes da OMS para os cuidados perinatais de mais qualidadeNovas diretrizes da OMS para os cuidados perinatais de mais qualidade
Novas diretrizes da OMS para os cuidados perinatais de mais qualidade
Prof. Marcus Renato de Carvalho
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Dr KHALID B.M
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
Shweta
 
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
GL Anaacs
 
micro teaching on communication m.sc nursing.pdf
micro teaching on communication m.sc nursing.pdfmicro teaching on communication m.sc nursing.pdf
micro teaching on communication m.sc nursing.pdf
Anurag Sharma
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Saeid Safari
 
Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...
Sujoy Dasgupta
 
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTSARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
Dr. Vinay Pareek
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
aljamhori teaching hospital
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Dr Jeenal Mistry
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
SumeraAhmad5
 
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptxPharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Dr. Rabia Inam Gandapore
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
MedicoseAcademics
 

Recently uploaded (20)

ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIONDACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
 
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness JourneyTom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
Tom Selleck Health: A Comprehensive Look at the Iconic Actor’s Wellness Journey
 
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.GawadHemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
 
Novas diretrizes da OMS para os cuidados perinatais de mais qualidade
Novas diretrizes da OMS para os cuidados perinatais de mais qualidadeNovas diretrizes da OMS para os cuidados perinatais de mais qualidade
Novas diretrizes da OMS para os cuidados perinatais de mais qualidade
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
 
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
 
micro teaching on communication m.sc nursing.pdf
micro teaching on communication m.sc nursing.pdfmicro teaching on communication m.sc nursing.pdf
micro teaching on communication m.sc nursing.pdf
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
 
Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...
 
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTSARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
 
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptxPharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
 

Whartons jelly review

  • 1. 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; 2 Graduate 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 Stem cells are defined by their unique capacity to self- renew, differentiate into one or more cell lineages and tissue types, and functionally repopulate a tissue in vivo [1]. On the basis of their ability to give rise to one or more cell types, known as potency, stem cells can be hierarchically classified as unipotent, multipotent, pluripotent and totipotent. Unipo- tent stem cells can produce only one cell type (e.g. neurons), whereas multipotent ones can differentiate into several cell types (e.g. hematopoietic stem cells, HSCs). An embryonic stem cell (ESC), derived from the inner cell mass (ICM) of the embryo is pluripotent as can differentiate into cells of the three germ layers, but not into trophoblastic cells, whereas the zygote is totipotent as it produces cells of embryonic as well as extraembryonic tissue. HSCs isolated from post-natal bone marrow (BM) and umbilical cord blood (UCB) are generally acknowledged as the prototype of multipotent adult tissue stem cells, being able to differentiate into all mature blood lineages as well as serially reconstitute hematopoiesis in lethally irradiated mice at the single cell level [2]. In the late 60s and early 70s, *Address correspondence to this author at the Department of Hematology, University Hospital of Heraklion, P.O. Box 1352, Heraklion, Crete, Greece; Tel: +30 2810 394629 ; Fax: +30 2810 394632 ; E-mail: pontikoglou@yahoo.com Friedenstein et al. demonstrated that a population of non- hematopoietic, plastic-adherent, fibroblast-like cells can also be isolated from BM [3]. These cells, which were later shown to be able to give rise to adipocytes, chondrocytes, osteocytes, smooth muscle cells, fibroblasts and hema- topoietic supportive stroma [3-6] and be self-renewing [7], are currently referred to as mesenchymal stem/stromal cells (MSCs) [4, 5, 8, 9]. So far MSC-like cells have been isolated from several tissues, such as skeletal muscle, adipose tissue, umbilical cord, synovium, dental pulp, amniotic fluid, as well as fetal blood, liver, BM, lung and heart [10-12]. Never- theless, these populations are not equivalent in terms of mul- tipotency, when in vivo assays are used [13]. The field of MSCs has witnessed considerable progress over the last decade. Due to the unraveling of many of their exceptional characteristics and the encouraging preclinical and clinical studies, it is now widely accepted that MSCs will provide a revolutionary advantage in the therapeutic intervention of various diseases or tissue damage in the near future. More than forty years since their initial description, BM-MSCs represent the most extensively studied population of adult MSCs and are still considered as the gold-standard for MSC clinical applications. Nevertheless it is now becom- ing increasingly clear that BM may not represent the most suitable source for MSC collection. Besides the invasive and painful isolation procedure, MSCs are present at very low frequency (approximately 0.001–0.01% of BM mononuclear 2212-3946/13 $58.00+.00 © 2013 Bentham Science Publishers
  • 2. 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 and quantitative properties decline with donor's age [15]. The low frequency implies that an extensive in vitro expan- sion of cells is usually required to obtain sufficient cell num- bers for therapeutic interventions, thereby enhancing the risk of epigenetic damages as well as viral and bacterial contami- nations. To circumvent these obstacles, much effort has been invested over the last decade in identifying alternative, more abundant and easily attainable sources of MSCs for thera- peutic use. In this regard the umbilical cord (UC), which is considered medical waste and is obtained with a painless, simple and safe procedure during delivery, has aroused much attention during the last years as a promising source for MSC isolation. MSCs have actually been found in both UC blood (UCB) as well as in UC tissues. Nevertheless, the scarcity of MSC in UCB (their frequency being much lower than in BM) and the lack of a robust and reproducible technique for harvesting and expanding MSCs from UCB units [16, 17] have hampered their use in clinical applications. On the other hand existing isolation protocols have constantly induced the harvest of large numbers of MSCs from UC-tissues with a hundred per cent success. UC-MSCs share many properties with adult BM-MSCs, but are generally considered as a more primitive population than the latter. In fact, while they possess several characteris- tics of ESCs, they do not form teratomas upon transplanta- tion [18] and their research does not raise ethical or legal issues. All these features open attractive perspectives for the propagation and engineering of UC-derived MSCs for cell- based therapies. This review will first attempt to give a summary of the biology of MSCs derived from Wharton's Jelly (WJ-MSCs), the most abundant region of UC and then discuss their potential application for the treatment of cancer and immune mediated disorders such as graft versus host disease (GVHD) and systemic lupus erythematosus (SLE). Finally, their putative role as feeder layers for ex vivo hema- topoietic stem cell (HSC) expansion will be pointed out. Human Umbilical Cord is a Rich Source of MSCs The human UC (Fig. 1) contains two arteries and a vein buried within a mucous or gelatinous connective tissue also known as the Wharton’s jelly (WJ), enclosed by a simple amniotic epithelium. WJ is largely made from proteoglycans and various types of collagen, forming a sponge-like tissue, within which stromal cells are embedded [19]. These cells, also referred to as umbilical cord matrix (UCM) cells are thought to be trapped into the WJ early in embryogenesis during their migration from the aortic-gonadotropin- mesonephric region to the fetal liver through the umbilical cord [20]. UCM cells have exceptional properties in that although they are bona fide MSCs [18, 21] as discussed be- low in detail, possessing similar properties with their adult BM counterparts, they also retain characteristics of primitive stem cells, like the expression of embryonic stem cell mark- ers [22, 23]. In line with these observations MSCs derived from WJ, hereafter referred to as "UC-MSCs", maintain their self-renewal capacity in vitro for a longer period of time, and exhibit a shorter doubling time and a broader pluripotency as compared to BM-MSCs [18]. These unique developmental and operational characteristics along with the fact that UC- MSCs can be easily isolated, extracorporeally without risks for the donor, from a tissue which is routinely discarded, highlight the advantages of UC-MSCs over ESCs and adult MSCs and explain the rapidly growing interest in this field of stem cell biology. The first report providing robust evidence that WJ stro- mal cells can be classified as MSCs was published in 2004 [24]. In that study, cells extracted from WJ, displayed a fi- broblast-like phenotype when expanded in vitro and were able to differentiate into osteoblasts, adipocytes and chon- drocytes upon appropriate stimulation [24]. Furthermore, akin to BM-MSCs they were shown to express high levels of CD29, CD44, CD51, CD73 and CD105, but lacked expres- sion of the hematopoietic markers CD34 and CD45 (Table 1) [24]. Subsequent reports have provided additional informa- 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. A Wha A U bili l iUmbilical arteries Perivascular Intervasc Amniotic epithelium arton’s Jelly Umbilical vein/ Umbilical cord blood Amniotic epithelium Subam cular nion
  • 3. 146 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al. tion on WJ stromal cell phenotype, demonstrating the ex- pression of various membrane molecules known to be asso- ciated with a MSC-like identity. These molecules include CD90, CD146, CD166 and ganglioside 2 (GD2) [8, 23, 25, 26]. Collectively, the morphology, immunophenotype and differentiation potential of WJ stromal cells fulfill the criteria for MSCs proposed by the International Society for Cellular Therapy (ISCT) [8]. Indeed, given that at present there is no specific marker to define UC-MSCs, the expression of the aforementioned set of membrane antigens combined with the demonstration of in vitro multi-lineage differentiation poten- tial is necessary to identify MSCs in UC-derived tissues UC-MSCs have been isolated from three relatively indis- tinct regions of the WJ: the perivascular, the intervascular and the subamnion zones (Fig. 1) [27]. Cells from these zones share a fibroblast-like morphology, a high proliferative rate and multipotent differentiation capacity. However a comprehensive head-to-head comparison between MSCs obtained from different regions of the UC has not been per- formed thus far, due to the great variation in cell isola- tion/culture protocols. Hence the question whether cells har- vested from different components of the UC represent differ- ent populations is still open. Nevertheless one study combin- ing both in situ and in vitro experiments [27] has demon- strated that MSCs derived from the perivascular region com- prise a distinct subpopulation than those derived from the intervascular and subamniotic parts of the UC. It has thus been reported that perivascular MSCs stain strongly for pan- cytokeratin as compared to intervascular and subamniotic counterparts and give rise in vitro to flat, wide cytoplasmic cells, named type 1 cells, which tend to diminish in number during protracted culture [27]. On the other hand intervascu- lar and subamniotic MSCs generate fibroblast-like cells per- sisting throughout culture (type 2 cells). In view of these findings it appears that UC-MSCs are actually type 2 cells, displaying adipogenic, chondrogenic, osteogenic and neu- ronal capacity, upon appropriate differentiation stimuli [27]. Isolation of MSCs from UC perivascular zone has been re- ported by other groups as well [28, 29]. These cells, termed as Human Umbilical Cord Perivascular Cells (HUCPVCs), have a colony forming unit (CFU) frequency of 1:333 [29] and differ in their growth characteristics from UC-MSCs (population doubling times ranging from 59.4±42.4h in pas- 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 population of UC-MSCs [27, 29]. Furthermore, HUCPVCs exhibit higher proliferative capacity and osteogenic potential as compared to BM-MSCs [28]. The phenotype of HUCPVCs shares many similarities with that of WJ-derived MSCs (Table 1). One significant difference though, is the high expression of CD146 by HUCPVCs. Interestingly this marker is either absent [28] or weakly expressed by MSCs derived form other regions of WJ [30]. Isolation of MSCs from Wharton's Jelly In recent years, several methods (Fig. 2) have been de- veloped for the isolation of MSCs from WJ. However, ex- perimental approaches vary considerably and in some cases harvested cells from UC tissue most likely consist of a het- erogeneous population, thereby biasing comparisons. With regards to protocols used to isolate UC-MSCs, some authors remove umbilical cord vessels, scrape off WJ with a scalpel and then treat the tissue enzymatically [24]. Others cut down UC to smaller segments, strip the vessels and then directly immerse the remaining UC to an enzymatic solution [25, 27, 31]. Interestingly, neither the enzymes used nor the incuba- tion period have been standardized thus far. Alternatively, instead of enzymatic digestion, several groups dissect the cord segment into very small pieces, with or without discard- ing the cord vessels and the segments are used as explants from which cultures are subsequently established [32-34]. The major advantage of this method is that it minimizes cell damage as it does not involve enzymatic treatment, however it should be noted that primary cultures generated in this way are quite heterogeneous [33]. UC-MSCs have also been iso- lated from WJ by simply cutting UC segments longitudinally and plating them with WJ side-down onto a plastic surface, once again without enzyme digestion or dissection [35]. Fi- nally, to collect HUCPVCs some groups remove blood ves- sels together with surrounding WJ, tie up the ends of each vessel into a loop and then immerse the vessel in enzyme, thereby isolating cells from the perivascular tissue [28, 29]. The question as to which is the optimal method for effi- cient isolation of a homogeneous and well defined popula- tion of UC-MSCs has not been answered thus far. However, unless an agreement is reached between laboratories regard- ing the isolation and ex vivo expansion of UC-MSCs, there will be no consensus on the characteristics and biological properties of the cells, which is a prerequisite for the proper design of preclinical studies, so as to pave the way for the potential use of UC-MSCs in clinical applications. Do differences in isolation procedures and ex vivo expan- sion protocols have an impact on the antigens expressed by UC-MSCs? This issue has not been properly investigated so far, yet a recent study [36] demonstrated that MSC markers CD73, CD90 and CD105 varied their expression patterns according to whether enzyme cocktails or explants were used for UC-MSC harvesting. A similar variation with respect to the isolation method was also noted in the expression of the ESC markers c-Kit and Oct-4 by UC-MSCs [36]. WJ-derived UC-MSCs may be Regarded as a Primitive Stromal Cell Population Porcine UC-MSCs have been reported to express the ESC markers Oct-4, Sox-2 and Nanog [37] and equine UC- MSCs express, apart from Oct-4, the embryonic markers SSEA4 and c-Kit [38]. In line with these findings, human UC-MSCs express Oct-4, Nanog, Sox-2, c-Kit, as well as Dnmt3b and Tert, albeit at lower levels as compared to ESCs [22, 34, 39]. Such a low expression of the aforementioned pluripotency markers may explain why UC-MSCs do not produce teratomas in vivo in immunodeficient mice and rats [22, 25]. In addition human UC-MSCs express the ESC markers Tra-1-60, Tra-1-81, SSEA-1 and SSEA-4 [22]. Many of these pluripotent stem cell markers retain their ex- pression for at least nine passages during ex vivo expansion. Comparative gene expression profiling between UC-MSCs and BM-MSCs demonstrated that the former express higher levels of Nanog, Dnmt3b and Gabrb3, and also
  • 4. 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] CD54 ICAM-1 (intercellular adhesion molecule-1) - [47] + [105] + [29] + [5] 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] CD106 VCAM-1 (Vascular cell adhesion molecule-1) - [49] + [61] - [29] + [107] + [5] CD117 C-kit receptor - [104] + [108] - [49] + [29] - [102] 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]
  • 5. 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] STRO-1 - [81] + [112] + [113] 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] Fig. (2). A scheme depicting the different approaches followed for the establishment of WJ-derived UC-MSC cultures (modified from [100]). Umbilical cord Cut tissue into very small pieces Dissection in small segments Explant culture Stromal cell culture Filter Removal of umbilical vessels Scrapping off mesenchymal tissue Enzymatic treatment
  • 6. 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 markers Brix, CD9, Gal, Kit and Rex1 [39]. The implication of these markers in the biology of UC-MSCs is currently unknown, yet their expression is thought to reflect the more primitive nature of UC-MSCs as compared to their adult counterparts within the BM. However, not all UC-derived MSCs express ESC markers. In fact, it has been demon- strated that primitive markers are expressed only by a subset of UC-MSCs [26, 40], thereby suggesting that these cells are rather hetereogeneous in terms of stemness. It would there- fore be very challenging to identify that specific marker which would permit the isolation of the primi- tive/multipotent MSCs from UC. Human UC-MSCs exhibit also higher expression of genes implicated in the phosphoinositide 3-kinase (PI3K)- AKT survival/proliferation pathway, including GAB2, NFKBIA, FOXO1A, YWHAB, FOXO3A, CDKN1A, MAP3K8, NFKB1, NOS3, DDR1 and TEK [41]. Further- more, the NF -B pathway has been reported to be more ac- tive in UC-MSCs than in BM-MSCs [41]. These findings together with the fact that human WJ-MSCs express the primitive stem cell marker TERT [25] explain, at least in part the observation that UC-MSCs have a shorter doubling time [27] and a more extensive ex vivo expansion capacity [18]. Multilineage in vitro Differentiation Potential of WJ- derived UC-MSCs Accumulating evidence suggests that UC-MSCs can be successfully induced to differentiate into mesodermal and non-mesodermal lineages. As mentioned above, several groups have established that UC-MSCs can generate os- teoblasts, adipocytes and chondrocytes in vitro [24, 27, 42, 43], thereby fulfilling one of the minimal criteria proposed by ISCT for MSCs [8]. However, lipid accumulation takes much longer in UC-MSCs and the adipocytes are less mature than those derived from BM-MSCs [27, 41]. In addition, UC-MSCs have been shown to possess weaker osteogenic [41] and chondrogenic [44, 45] potential as compared to BM-MSCs. UC-MSCs have also been reported to successfully differ- entiate into endothelial cells [43, 46] and actually demon- strate higher endothelial differentiation potential when com- pared to BM-MSCs [43]. In accordance with this observation non-differentiated UC-MSCs exhibit higher expression of endothelial associated genes including FLT1, GATA4, GATA6, ISL1, LAMA1, SOX17 and SERPINA1 [39]. One group has suggested, that UC-MSCs can give rise to skeletal muscle cells when cultured in the appropriate me- dium and that non-differentiated UC-MSCs may display myogenic potential in vivo [47]. Differentiation of UC-MSCs to cardiomyocytes has also been reported but is still contro- versial [24, 31, 48-50]. For instance, one group [24] demon- strated that UC-MSCs could be appropriately induced to form cells demonstrating cardiomyocyte morphology and expressing cardiac markers, including N-cadherin and car- diac troponin. Another team [49], however, was unable to detect cardiomyocyte marker expression after in vitro induc- tion of MSCs isolated from WJ and perivascular tissues. De- finitive evidence that UC-MSC-derived cardiomyocytes rep- resent functional and terminally differentiated cells is still scarce. To our knowledge, only one group has thus far re- ported that UC-MSC-derived cardiomyocytes exhibit slight spontaneous beating after 21 days of induction [31]. Finally, several groups have shown that UC-MSCs can acquire morphological and biochemical properties of neural cells when exposed to appropriate differentiation media [27, 32, 51, 52]. Mitchell et al. [32] were the first who observed UC-MSC neuronal differentiation, following a complex and multi-step neuronal induction process. Differentiation was assessed by changes in morphology and expression of neu- ron specific enolase, bIII-tubulin, neurofilament and tyrosin hydroxylase. Subsequent studies have further corroborated these findings. It has also been suggested that UC-MSCs may be more primed for neuronal differentiation as com- pared to BM-MSCs, since they express, higher levels of neu- ronal-associated genes including NESTIN, GFAP and SEM3A [39] in a non–differentiated state. However, it should be kept in mind that definitive evidence for electro- physiological activity of UC-MSC-derived neurons is still missing. Therefore the issue whether the neuronal-like phe- notype of UC-MSCs reflects a genuine trans-differentiation is still pending. Recent studies suggest that UC-MSCs can also differen- tiate into endodermal lineages (reviewed in [53]). In this context, Campard et al. [54] reported that non-differentiated UCM cells constitutively expressed markers of hepatic line- age, such as albumin, alpha-fetoprotein, cytokeratin-19, con- nexin-32, and dipeptidyl peptidase IV. After in vitro hepatic induction, cells exhibited hepatocyte-like morphology and upregulated the expression of certain hepatocyte-associated markers, but lacked important functional properties of ma- ture liver cells, thereby questioning whether UCM cells have actually undergone hepatocyte maturation [54]. Other groups have also claimed that UC-MSCs can be induced to form isle-like cells [55, 56] (reviewed in [57]), able to express pancreatic associated genes and release insulin in response to physiological glucose levels [56]. Interestingly, UC-MSCs have been reported to have a higher pancreatic differentia- tion potential than their BM counterparts [55]. The Immunomodulatory Properties of UC-MSCs Apart from the multilineage potential, UC-MSCs possess various immunoregulatory properties, rendering them as appealing candidates for cell based therapies [58]. More pre- cisely UC-MSCs have been found to exhibit very low ex- pression of HLA class I, and lack the expression of HLA-DR [59, 60]. Notably, in comparison to BM-MSCs derived from aged donors, UC-MSCs express significantly lower levels of HLA-I, and exhibit significantly higher intracellular concen- trations of the immunosuppressive molecule HLA-G [59]. These findings together with the observations that UC-MSCs do not express the co-stimulatory molecules CD40, CD80 and CD86 [59, s61], and do not elicit proliferative responses from allogeneic T cells in vitro [61], suggest that UC-MSCs may be characterized by inherently low immunogenicity. However, another study suggests that UC-MSCs may not be as immune-privileged as thought, as they can trigger weak allogeneic immune activation in vitro and will eventually
  • 7. 150 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al. undergo rejection following xenogeneic transplantation [59]. Similar findings have been reported for BM-MSCs as well. Interestingly the latter have been shown to be much more immunocompetent compared to UC-MSCs [59]. Several studies have been published on the immunosup- pressive potential of UC-MSCs on T lymphocytes [59, 60, 62-64]. One group reported that UC-MSCs target CD4+ and CD8+ T subpopulations equally and are able to inhibit T-cell proliferation regardless of the stimulus used to activate T- cells [63]. Furthermore, UC-MSC-mediated inhibitory ef- fects were higher than with BM-MSCs. In accordance with previous studies suggesting that the immunomodulatory ef- fect of MSCs on T cells may be mediated, at least in part, by soluble factors (reviewed in [65]), such as hepatocyte growth factor (HGF), prostaglandin E2 (PGE2), transforming growth factor- 1 (TGF- 1) and indoleamine 2,3-dioxygenase (IDO), the authors assessed whether any of these molecules may be implicated in UC-MSC immunoregulation as well. In their hands immune suppression exerted by both UC- and BM- MSCs was mediated by cycloxygenase (COX) 1 and COX2 enzymes, regulating PGE2 synthesis, and by the production of PGE2 per se, whereas HGF did not seem to be actively implicated. These findings were further corroborated by an- other study [64] which additionally showed that while UC- MSCs intrinsically secrete very low levels of PGE2, its pro- duction is upregulated when cells are co-cultured in the pres- ence of activated human peripheral blood mononuclear cells (hPBMCs). The authors attributed the PGE2 stimulation to inflammatory cytokines (including IFN- and interleukin-1 ) produced by hPBMCs [64]. Interestingly, several studies [59, 62, 66] have reported that UC-MSC immune characteristics may be modified fol- lowing exposure to inflammatory cytokines and/or activated PBMCs/T-cells so as to display immunosupressive proper- ties, a process described by the term licensing or priming. In this context, Deuse et al. [59] reported that IFN- at doses below 50ng/ml upregulated HLA-DR and HLA-I expression, thereby increasing UC-MSC immunogenicity. IFN- treat- ment also induced tolerogenicity by increasing intracellular HLA-G expression, TGF- and IL-10 release, and stimulated the production of IDO [59]. Higher doses of IFN- however enhanced UC-MSC immunosuppressive phenotype by downregulating HLA-DR expression and further increasing IDO. These results suggest that appropriate priming may be crucial for the fine adjustment of MSC characteristics, ren- dering them suitable for a given therapeutic application. The immunomodulatory effects of MSCs are not only apparent in T lymphocytes but also concern B lymphocytes. To our knowledge however, the impact of UC-MSCs on B cells has been poorly investigated and undoubtedly awaits further elucidation, as the information that will be gathered is expected to provide crucial clues for the introduction of WJ- MSCs into the clinic. In the single study reported until today [67] UC-MSCs were found to inhibit B cell proliferation, differentiation and antibody production in vitro. This sup- pressive effect is mediated, at least in part by soluble factors, which may downregulate Blimp-1 and upregualate PAX-5, two master regulators of B cell differentiation [67]. Further- more UC-MSCs have been demonstrated to inhibit Akt and p38 MAPK phosphorylation. Interestingly, these signal transduction pathways are implicated in the regulation of B cell proliferation and differentiation [67]. The immunoregulatory potential of HUCPVCs has also been assessed in a recent study [68]. More precisely it has been demonstrated that when HUCPVCs are co-cultured with resting or stimulated allogeneic lymphocytes they fail to induce their proliferation. Furthermore, HUCPVCs have been found to reduce in vitro alloreactivity in mixed lym- phocyte reactions. Notably, the immunosuppressive effect of the cells is exerted, at least in part via soluble factors [68]. UC-MSCs as Feeder Layers for ex vivo Umbilical Cord Blood Expansion Despite the fact that BM has been the most extensively studied source for HSC transplantation, the importance of UCB as an alternative source of transplantable HSCs has been widely acknowledged since the first successful UCB transplant in 1988 [69]. From a practical point of view UCB transplantation has several advantages over BM, including the non-invasiveness of the procedure, the ease of procure- ment, the reduced risk of transmissible infections and the availability for immediate use [70]. In addition, UCB grafts allow for a greater tolerable HLA disparity with a decreased frequency of acute or chronic graft versus host disease (GVHD) as compared to BM grafts [70]. Finally UCB con- tains a higher proportion of immature and committed hema- topoietic progenitor cells (HPCs) in comparison to adult BM [71]. However UCB transplantation has also some significant restrictions, limiting its broad applicability. The most sig- nificant limiting factor of UCB use is probably the low total cell dose harvested from a UCB unit, which in turn is associ- ated with delayed neutrophil and platelet engraftment and higher engraftment failure [72]. For this reason UCB trans- plantation remains significantly more successful in children [73], whereas poor outcomes have been reported in patients >45 kgr who receive only one unit of UCB [74]. To circumvent these obstacles, so as to improve out- comes and extend the application of UCB transplantation to older and/or larger patients, several groups have explored the possibility of in vivo expansion of HSCs from UCB units. Various protocols are under investigation over the last 30 years, including those based on cultures on feeder layers. Among the different cell types that have been used as stro- mal support for UCB expansion, the most extensively stud- ied thus far are BM- MSCs. The rationale behind the use of MSCs as feeder cells for UCB expansion lies on a large body of evidence, clearly demonstrating the BM-MSC promoting role on hematopoie- sis (reviewed in [65]). More precisely BM-MSCs have been shown in vivo to enhance HSC engraftment and repopula- tion, as evidenced by the observation that co-transplantation of human HSCs and MSCs results in increased chimerism and/or hematopoietic recovery, in both animal models and humans [65, 75]. On the other hand, MSCs have been shown in vitro to maintain long-term culture initiating cells (LT- CICs) and expand lineage-specific colony forming units from CD34+ marrow cells in long-term BM cultures (re- viewed in [65]). Even though the exact mechanisms that me-
  • 8. 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 been elucidated, it is now widely accepted that MSCs supply HSCs and their progeny with signals for survival, prolifera- tion and differentiation through direct cell-to-cell contact and/or production of cytokines, adhesion molecules and ex- tracellular matrix proteins (reviewed in [65]). These observations have provided the theoretical back- ground for the investigation of the potential use of MSCs as scaffolds for the ex vivo expansion of HSCs. Based on the hypothesis that MSCs could provide many of the molecular cues of the hematopoietic microenvironment necessary to favor HSC self-renewal and regulate their proliferation, CB hematopoetic stem/progenitor cells have been co-cultured on BM-MSC preformed layers in a medium containing (FBS) and various combinations of growth factors including G- CSF, stem cell factor (SCF), thrombopoietin (TPO), Flt3- ligand or fibroblast growth factor-1 (FGF-1) [76]. Using this ex vivo model several groups have demonstrated an increase in the number of total nucleated cells (TNC), clonogenic cells or CD34+ cells [77, 78]. These findings were further corroborated in a recent study [79], where CB CD34+ cells were co-cultured with BM-MSCs in a serum free medium supplemented with SCF, TPO and FGF-1. BM-MSCs were also shown to maintain the primitive phenotype of CB hema- topoietic stem and progenitor cells even after massive expan- sion [79]. However co-culture with BM-MSCs did not result in a significant expansion of long-term repopulating cells. A clinical trial is underway at M.D. Anderson Cancer Center combining an unmanipulated UCB unit with an expanded UCB unit for 14 days in the presence of a cytokine cocktail on layer of family member donor-derived BM-MSCs or "off- the-self" BM-MSCs (Angioblast Systems Inc.). Preliminary results suggest a beneficial role for these stromal cell-based HSC expansion [80]. Given the promising role of BM-MSCs in supporting ex vivo UCB expansion and since UC-MSCs share many char- acteristics with their BM counterparts, but in contrast to the latter can be readily and painlessly harvested from UC, they soon emerged as attractive candidates for HSC expansion. In this regard, the first report demonstrating that akin to BM- MSCs, UC-MSCs can also support hematopoiesis was pub- lished in 2007 [81]. In that study the authors showed that UC-MSCs secrete significantly higher amounts of several important cytokines and hematopoietic growth factors in- cluding G-CSF, GM-CSF, LIF, IL-1 , L-6, IL-8 and IL-11, compared to BM-MSCs. Furthemore co-culture of UC- MSCs with UCB CD34+ cells significantly increased hema- topoietic colony formation as compared to clonogenic pro- genitor cell assays performed in the absence of MSCs [81]. Finally, in agreement with previous reports demonstrating that co-transplantation of BM-MSCs with HSCs can acceler- ate hematopoietic engraftment, co-injection of UC-MSCs with UCB cells or CD34+ cells in a NOD/SCID mouse model accelerated human HSC recovery. The authors as- sumed that the improved engraftment might be attributed to the presence of UC- MSCs within the BM of the recipient animals and also to the production of hematopoietic cytoki- nes by these cells. Subsequent reports have confirmed the capacity of UC-MSCs and UC-MSC conditioned medium to support the growth and maintenance of UCB hematopoietic stem/progenitor cells in long term culture [82, 83], thereby establishing an ex vivo model that could enable the study of the interactions between HSCs and their supportive micro- environment within the UC. However, whether UC MSC- based HSC expansion will have possible future clinical util- ity is currently unknown and awaits further clarification. UC-MSCs for GVHD Treatment Graft versus host disease (GVHD) is one of the most severe complications of allogeneic BM transplantation. First- line treatment with corticosteroids is the gold standard for this T-cell mediated process, however second line treatment for steroid-resistant GVHD has not been clearly established thus far and the outcome is poor, with one year overall sur- vival (OS) of only 30% [84, 85]. The in vitro and in vivo immunomodulatory properties of BM-MSCs have provided the theoretical background for the use of these cells for treatment of acute steroid-resistant GVHD. Several reports have been published thus far (re- viewed in [86]) suggesting that clinical responses can actu- ally be attained in a significant number of patients, especially in children. BM-MSCs have also been used for the treatment of chronic GVHD showing likewise promising results [86]. To our knowledge, there has been only one recent study clinically testing UC- MSCs for GVHD [87]. In that particu- lar report third-party culture expanded UC- MSCs were transplanted in 2 children with severe steroid-resistant acute GVHD. The first patient received three infusions of UC- MSCs. Each time acute GVHD improved significantly and eventually disappeared. The second patient received only one infusion of UC-MSCs that led to complete remission of acute GVHD. No child experienced any adverse events what so ever and both children were well and remained off immu- nosuppressive therapy at the time the study was published. The authors concluded that UC-MSCs may be considered as a feasible, safe and effective treatment modality for acute GVHD [87]. These encouraging results need obviously to be confirmed by larger trials, which will include adult patients as well. Furthermore, additional studies are needed in order to optimize the use of UC-MSCs in treating GVHD. For ex- ample, it is currently unknown which is the optimal UC- MSC dose and schedule, or whether the use of platelet- lysate, or other substitutes of the standard FBS-enriched me- dium (so as to comply with regulatory guidelines for the production of MSCs for clinical use, which recommend avoiding the use of animal derivatives) might influence the therapeutic effect of UC-MSCs. UC-MSCs Support the Expansion of Natural Killer Cells As previously mentioned a major toxicity associated with Bone Marrow Transplantation (BMT) is GVHD, an attack by the T cells in the graft against the recipient organs includ- ing the skin, gastrointestinal tract, liver, and eyes [88]. Ag- gressive depletion of T cells in the allogeneic graft can markedly reduce the incidence of acute GVHD, however, this is associated with significant drawbacks, including re- duction of the graft versus tumor effect of the allogeneic- BMT [89]. Limiting therefore GVHD while maintaining the
  • 9. 152 Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 Batsali et al. antitumor effect, would be critical to improve transplant out- come [90]. Given that natural killer (NK) cells have been found to control leukemia relapse while protecting patients from GVHD [91, 92], the possibility of expanding and po- tentially using these cells for adoptive immunotherapy ap- pears an attractive approach in the setting of allogeneic BMT. Provided that UCB is rich in NK cell progenitors and since UC-MSCs can produce significant amounts of cytoki- nes, Boissel et al. [93] hypothesized that a feeder layer of UC-MSCs might actually effectively support ex vivo expan- sion of UCB-NK cells. Indeed when CD3-depleted UCB mononuclear cells were cocultured with UC-MSCs and a cytokine cocktail, the proportion of CD3- CD56+ NK cells increased significantly as compared to cell cultured with cytokines only. Interestingly direct contact between UCB mononuclear cells and UC-MSCs resulted in a significantly greater NK cell expansion. Notably, expanded NK cells maintained an elevated cytotoxic profile [93]. The possibility of further optimizing this experimental protocol should be evaluated in additional studies and of course the cytotoxicity of the expanded NK cells awaits assessment in animal mod- els. UC-MSC Transplantation in Systemic Lupus Erythema- tosus To our knowledge, apart from GVHD, in vivo therapeutic efficacy of UC- MSC transplantation has also been clearly illustrated in the setting of systemic lupus erythematosus (SLE). The theoretical background for assessing the clinical impact of UC-MSCs in patients with this autoimmune disor- der has been provided by the immunomodulatory properties of UC-MSCs and also by the beneficial effects of BM- MSC administration in patients with treatment-refractory SLE [94]. In this context, it has recently been reported that trans- plantation of UC-MSCs in patients with severe and refrac- tory SLE results in clinical and serologic improvement and is not associated with mortality or other treatment-related ad- verse events [95]. Even though the mechanisms mediating the therapeutic effect of UC-MSCs in SLE patients remain unclear, it has been suggested that disease remission may be associated with an increase in the proportion of Tregs in pe- ripheral blood along with a decrease in IL-4 serum levels, probably resulting in inhibition of humoral activity [95]. Another recent study has demonstrated that UC-MSCs can also provide clinical benefit in diffuse alveolar hemorrhage, a rare complication of SLE with high clinical mortality [96]. These findings suggest that UC-MSC transplantation may hold promise as a treatment modality in selected SLE cases. However, the efficacy and safety of this strategy need to be further validated in larger clinical trials with longer periods of follow-up. Therapeutic Potential of UC-MSCs in Cancer An emerging clinical application of UC-MSCs that re- cently gained much attention is their use as cellular therapy 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 severe combined immunodeficiency (SCID) mice, bearing human breast carcinoma tumors in their lungs [97]. Inter- estingly, UC-MSCs were only found in the lungs and not in other tissues. This combined approach resulted in significant metastatic tumor reduction, much greater than compared to each treatment alone. A more recent study has corroborated the ability of UC-MSCs to reduce the growth of human breast carcinoma cells both in vitro as well as in vivo, in a mouse xenograft model [98]. Notably, the antitumor proper- ties of UC-MSCs do not seem to be specific only to breast cancer cells, but have also been demonstrated in osteosar- coma and ovarian carcinoma cell lines in vitro [99]. The mechanisms mediating UC-MSC anticancer effects are still under investigation. However, it has been suggested that UC- MSCs exert their antitumor activity both via cell-to-cell con- tact and humoral factors, through induction of apoptosis, cell cycle arrest and autophagy [97, 99]. Taken together, the aforementioned preliminary data imply that UC-MSCs may hold promise as a therapeutic modality in cancer and high- light the need for the development of additional well- designed pre-clinical studies so as to properly investigate the potential of this cell-based approach. CONCLUSION Within recent years considerable interest has been given to the exciting properties of UC-MSCs, derived from the WJ, because of their primitive nature as compared to BM-MSCs (the prototype of adult MSCs), their multilineage potential across germ layers, the extensive in vitro proliferation and their immunosuppressive capacity. These characteristics along with the ease by which UC-MSCs are isolated from a tissue with unlimited availability, have generated much en- thusiasm regarding their potential applications in cell-based therapies. This expectation is being supported by both pre- and clinical data. In particular, in this review we reported on the promising results obtained in recent studies regarding the use of UC-MSCs for the treatment of cancer and immune mediated disorders, such as graft versus host disease and systemic lupus erythematosus and discussed UC-MSC role as feeder layers for ex vivo HSC expansion. Nevertheless much work has to be done so as to fully unravel the biologic features of UC-MSCs and especially those distinguishing them from adult MSCs. Furthermore, properly designed comparative studies need to be undertaken in the near future so as to clearly identify and characterize MSC subpopula- tions originating from different regions of WJ, in order to answer the long-standing question: whether different ana- tomical locations within WJ are associated with distinct qualitative and quantitative MSC properties. Notably, avail- able global expression technologies, like gene expression and comparative genomic hybridization (CGH) microarrays, proteome and secretome profiles, methylation mapping and ChIP-on-ChiP technologies are anticipated to contribute sig- nificantly to the better understanding of UC-MSC biology. It is anticipated that the data obtained by these studies will not only provide the theoretical background for the eventual in- troduction of UC-MSCs in the clinic, but would also be ma- jor contribution to stem cell biology in general. CONFLICT OF INTEREST The authors confirm that this article content has no con- flicts of interest.
  • 10. Mesenchymal Stem Cells Derived from Wharton's Jelly Current Stem Cell Research & Therapy, 2013, Vol. 8, No. 2 153 ACKNOWLEDGEMENTS Declared none. REFERENCES [1] Weissman IL, Anderson DJ, Gage F. Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiati- ons. Annu Rev Cell Dev Biol 2001; 17: 387-403. [2] Kondo M, Wagers AJ, Manz MG, et al. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu Rev Immunol 2003; 21: 759-806. [3] Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation 1974; 12: 331-40. [4] Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997; 276: 71-4. [5] Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284: 143-7. [6] Delorme B, Ringe J, Pontikoglou C, et al. Specific lineage-priming of bone marrow mesenchymal stem cells provides the molecular framework for their plasticity. Stem Cells 2009; 12: 1142-51. [7] Sacchetti B, Funari A, Michienzi S, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007; 131: 324-36. [8] Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for cellular therapy position statement. Cytotherapy 2006; 8: 315-7. [9] Caplan AI. Mesenchymal stem cells. J Orthop Res 1991; 9(5): 641- 50. [10] da Silva Meirelles L Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006; 119: 2204-13. [11] Anzalone R, Corrao S, Lo Iacono M, et al. Isolation and Characterization of CD276+/HLA-E+ Human Subendocardial Mesenchymal Stem Cells from Chronic Heart Failure Patients: Analysis of Differentiative Potential and Immunomodulatory Markers Expression. Stem Cells Dev 2013; 22: 1-17. [12] Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair--current views. Stem Cells 2007; 25: 2896- 902. [13] Bianco P, Robey PG, Saggio I, Riminucci M. "Mesenchymal" stem cells in human bone marrow (skeletal stem cells): a critical discussion of their nature, identity, and significance in incurable skeletal disease. Hum Gene Ther 2010; 21: 1057-66. [14] Castro-Malaspina H, Gay RE, Resnick G, et al. Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. Blood 1980; 56: 289-301. [15] Mueller SM, Glowacki J. Age-related decline in the osteogenic potential of human bone marrow cells cultured in three- dimensional collagen sponges. J Cell Biochem 2001; 82: 583-90. [16] Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B, Hows JM. Adult bone marrow is a rich source of human mesenchymal stem cells but umbilical cord and mobilized blood adult blood are not. Br J Haematol 2003; 121: 368-74. [17] Perdikogianni C, Dimitriou H, Stiakaki E, Martimianaki G, Kalmanti M. Could cord blood be a source of mesenchymal stromal cells for clinical use? Cytotherapy 2008; 10: 452-9. [18] Troyer DL, Weiss ML. Concise review: Wharton's jelly-derived cells are a primitive stromal cell population. Stem Cells 2008; 26: 591-9. [19] Nanaev AK, Kohnen G, Milovanov AP, Domogatsky SP, Kaufmann P. Stromal differentiation and architecture of the human umbilical cord. Placenta 1997; 18: 53-64. [20] Wang XY, Lan Y, He WY, et al. Identification of mesenchymal stem cells in aorta-gonad-mesonephros and yolk sac of human embryos. Blood 2008; 111: 2436-43. [21] Can A, Karahuseyinoglu S. Concise review: human umbilical cord Stroma with Regard to the source of fetus-derived stem cells. Stem Cells 2007; 25: 2886-95. [22] Fong CY, Richards M, Manasi N, Biswas A, Bongso A. Comparative growth behaviour and characterization of stem cells from human Wharton's jelly. Reprod Biomed Online 2007; 15: 708-18. [23] Fong CY, Chak LL, Biswas A,et al. Human Wharton's jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev 20011; 7: 1-16. [24] Wang HS, Hung SC, Peng ST, et al. Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells 2004; 22: 1330-7. [25] Weiss ML, Medicetty S, Bledsoe AR, et al. Human umbilical cord matrix stem cells: preliminary characterization and effect of transplantation in a rodent model of Parkinson's disease. Stem Cells 2006; 24: 781-92. [26] Xu J, Liao W, Gu D, Liang L, et al. Neural ganglioside GD2 identifies a subpopulation of mesenchymal stem cells in umbilical cord. Cell Physiol Biochem 2009; 23: 415-24. [27] Karahuseyinoglu S, Cinar O, Kilic E, et al. Biology of stem cells in human umbilical cord stroma: in situ and in vitro surveys. Stem Cells 2007; 25: 319-31. [28] Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells 2007; 25: 1384-92. [29] Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE. Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells 2005; 23: 220-9. [30] Moodley Y, Atienza D, Manuelpillai U, et al. Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol 2009; 175: 303-13. [31] Pereira WC, Khushnooma I, Madkaikar M, Ghosh K. Reproducible methodology for the isolation of mesenchymal stem cells from human umbilical cord and its potential for cardiomyocyte generation. J Tissue Eng Regen Med 2008; 2: 394-9. [32] Mitchell KE, Weiss ML, Mitchell BM, et al. Matrix cells from Wharton's jelly form neurons and glia. Stem Cells 2003; 21: 50-60. [33] Majore I, Moretti P, Stahl F, Hass R, Kasper C. Growth and differentiation properties of mesenchymal stromal cell populations derived from whole human umbilical cord. Stem Cell Rev 2011; 7: 17-31. [34] La Rocca G, Anzalone R, Corrao S, et al. Isolation and characterization of Oct-4+/HLA-G+ mesenchymal stem cells from human umbilical cord matrix: differentiation potential and detection of new markers. Histochem Cell Biol 2009; 131: 267-82. [35] De Bruyn C, Najar M, Raicevic G, et al. A rapid, simple, and reproducible method for the isolation of mesenchymal stromal cells from Wharton's jelly without enzymatic treatment. Stem Cells Dev 2011; 20: 547-57. [36] Salehinejad P, Alitheen NB, Ali AM, et al. Comparison of different methods for the isolation of mesenchymal stem cells from human umbilical cord Wharton's jelly. In vitro Cell Dev Biol Anim 2012; 48: 75-83. [37] Carlin R, Davis D, Weiss M, Schultz B, Troyer D. Expression of early transcription factors Oct-4, Sox-2 and Nanog by porcine umbilical cord (PUC) matrix cells. Reprod Biol Endocrinol 2006; 4: 8. [38] Hoynowski SM, Fry MM, Gardner BM, et al. Characterization and differentiation of equine umbilical cord-derived matrix cells. Biochem Biophys Res Commun 2007; 362: 347-53. [39] Nekanti U, Rao VB, Bahirvani AG, Jan M, Totey S, Ta M. Long- term expansion and pluripotent marker array analysis of Wharton's jelly-derived mesenchymal stem cells. Stem Cells Dev 2010; 19: 117-30. [40] Zhang ZY, Teoh SH, Chong MS, et al. Superior osteogenic capacity for bone tissue engineering of fetal compared with perinatal and adult mesenchymal stem cells. Stem Cells 2009; 27: 126-37. [41] Hsieh JY, Fu YS, Chang SJ, Tsuang YH, Wang HW. Functional module analysis reveals differential osteogenic and stemness potentials in human mesenchymal stem cells from bone marrow and Wharton's jelly of umbilical cord. Stem Cells Dev 2010; 19: 1895-910. [42] Huang YC, Parolini O, La RG, Deng L. Umbilical cord versus bone marrow-derived mesenchymal stromal cells. Stem Cells Dev 2012; 21: 2900-3.
  • 11. 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 Wharton's jelly-derived mesenchymal stem cells in comparison with bone marrow-derived mesenchymal stem cells. Exp Hematol 2009; 37: 629-40. [44] Lo Iacono M, Anzalone R, Corrao S, et al. Perinatal and Wharton's Jelly-Derived Mesenchymal Stem Cells in Cartilage Regenerative Medicine and Tissue Engineering Strategies. Open Tissue Eng Regen Med J 2011; 4: 72-81. [45] Wang L, Tran I, Seshareddy K, Weiss ML, Detamore MS. A comparison of human bone marrow-derived mesenchymal stem cells and human umbilical cord-derived mesenchymal stromal cells for cartilage tissue engineering. Tissue Eng Part A 2009; 15: 2259- 66. [46] Wu KH, Zhou B, Lu SH, et al. In vitro and in vivo differentiation of human umbilical cord derived stem cells into endothelial cells. J Cell Biochem 2007; 100: 608-16. [47] Conconi MT, Burra P, Di LR, et al. CD105(+) cells from Wharton's jelly show in vitro and in vivo myogenic differentiative potential. Int J Mol Med 2006; 18: 1089-96. [48] Zhao Z, Chen Z, Zhao X, et al. Sphingosine-1-phosphate promotes the differentiation of human umbilical cord mesenchymal stem cells into cardiomyocytes under the designated culturing conditions. J Biomed Sci 2011; 18: 37. [49] Martin-Rendon E, Sweeney D, Lu F, Girdlestone J, Navarrete C, Watt SM. 5-Azacytidine-treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies. Vox Sang 2008; 95: 137-48. [50] Qian Q, Qian H, Zhang X, et al. 5-Azacytidine induces cardiac differentiation of human umbilical cord-derived mesenchymal stem cells by activating extracellular regulated kinase. Stem Cells Dev 2012; 21: 67-75. [51] Ma L, Feng XY, Cui BL, et al. Human umbilical cord Wharton's Jelly-derived mesenchymal stem cells differentiation into nervelike cells. Chin Med J 2005; 118: 1987-93. [52] Fu YS, Cheng YC, Lin MY, et al. Conversion of human umbilical cord mesenchymal stem cells in Wharton's jelly to dopaminergic neurons in vitro: potential therapeutic application for Parkinsonism. Stem Cells 2006; 24: 115-24. [53] Anzalone R, Lo Iacono M, Corrao S, et al. New emerging potentials for human Wharton's jelly mesenchymal stem cells: immunological features and hepatocyte-like differentiative capacity. Stem Cells Dev 2010; 19: 423-38. [54] Campard D, Lysy PA, Najimi M, Sokal EM. Native umbilical cord matrix stem cells express hepatic markers and differentiate into hepatocyte-like cells. Gastroenterology 2008; 134: 833-48. [55] Wu LF, Wang NN, Liu YS, Wei X. Differentiation of Wharton's jelly primitive stromal cells into insulin-producing cells in comparison with bone marrow mesenchymal stem cells.Tissue Eng Part A 2009; 15: 2865-73. [56] Chao KC, Chao KF, Fu YS, Liu SH. Islet-like clusters derived from mesenchymal stem cells in Wharton's Jelly of the human umbilical cord for transplantation to control type 1 diabetes. PLoS One 2008; 3: e1451. [57] Anzalone R, Lo Iacono M, Loria T, et al. Wharton's Jelly Mesenchymal Stem Cells as Candidates for Beta Cells Regeneration: Extending the Differentiative and Immunomodulatory Benefits of Adult Mesenchymal Stem Cells for the Treatment of Type 1 Diabetes. Stem Cell Reviews and Reports 2011; 7: 342-63. [58] La Rocca G. Connecting the Dots: The Promises of Wharton's Jelly Mesenchymal Stem Cells for Tissue Repair and Regeneration. The Open Tissue Engineering and Regenerative Medicine Journal 2011; 12: 3-5. [59] Deuse T, Stubbendorff M, Tang-Quan K, et al. Immunogenicity and immunomodulatory properties of umbilical cord lining mesenchymal stem cells. Cell Transplant 2011; 20: 655-67. [60] Zhou C, Yang B, Tian Y, et al. Immunomodulatory effect of human umbilical cord Wharton's jelly-derived mesenchymal stem cells on lymphocytes. Cell Immunol 2011; 272: 33-8. [61] Weiss ML, Anderson C, Medicetty S, et al. Immune properties of human umbilical cord Wharton's jelly-derived cells. Stem Cells 2008; 26: 2865-74. [62] Yoo KH, Jang IK, Lee MW, et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cell Immunol 2009; 259: 150-6. [63] Najar M, Raicevic G, Boufker HI, et al. Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: Combined comparison of adipose tissue, Wharton's Jelly and bone marrow sources. Cell Immunol 2010; 264: 171-9. [64] Chen K, Wang D, Du WT, et al. Human umbilical cord mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through a PGE2-dependent mechanism. Clin Immunol 2010; 135: 448-58. [65] Pontikoglou C, Deschaseaux F, Sensebe L, Papadaki HA. Bone Marrow Mesenchymal Stem Cells: Biological Properties and Their Role in Hematopoiesis and Hematopoietic Stem Cell Transplantation. Stem Cell Rev 2011; 7: 569-89. [66] Valencic E, Piscianz E, Andolina M, Ventura A, Tommasini A. The immunosuppressive effect of Wharton's jelly stromal cells depends on the timing of their licensing and on lymphocyte activation. Cytotherapy 2010; 12: 154-60. [67] Che N, Li X, Zhou S, et al. Umbilical cord mesenchymal stem cells suppress B-cell proliferation and differentiation. Cell Immunol 2012; 274: 46-53. [68] Ennis J, Gotherstrom C, Le Blanc K, Davies JE. In vitro immunologic properties of human umbilical cord perivascular cells. Cytotherapy 2008; 10: 174-81. [69] Gluckman E, Broxmeyer HA, Auerbach AD, et al. Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling. N Engl J Med 1989; 321: 1174-8. [70] Gluckman E. Current status of umbilical cord blood hematopoietic stem cell transplantation. Exp Hematol 2000; 28: 1197-205. [71] Mayani H, Lansdorp PM. Biology of human umbilical cord blood- derived hematopoietic stem/progenitor cells. Stem Cells 1998; 16: 153-65. [72] Migliaccio AR, Adamson JW, Stevens CE, Dobrila NL, Carrier CM, Rubinstein P. Cell dose and speed of engraftment in placental/umbilical cord blood transplantation: graft progenitor cell content is a better predictor than nucleated cell quantity. Blood 2000; 96: 2717-22. [73] Gluckman E, Rocha V, Arcese W, et al. Factors associated with outcomes of unrelated cord blood transplant: guidelines for donor choice. Exp Hematol 2004; 32: 397-407. [74] Gluckman E, Rocha V, Boyer-Chammard A, et al. Outcome of cord-blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med 1997; 337: 373-81. [75] Almeida-Porada G, Flake AW, Glimp HA, Zanjani ED. Cotransplantation of stroma results in enhancement of engraftment and early expression of donor hematopoietic stem cells in utero. Exp Hematol 1999; 27: 1569-75. [76] Tung SS, Parmar S, Robinson SN, De LM, Shpall EJ. Ex vivo expansion of umbilical cord blood for transplantation. Best Pract Res Clin Haematol 2010;23:245-57. [77] McNiece I, Harrington J, Turney J, Kellner J, Shpall EJ. Ex vivo expansion of cord blood mononuclear cells on mesenchymal stem cells. Cytotherapy 2004; 6: 311-7. [78] Robinson SN, Ng J, Niu T, et al. Superior ex vivo cord blood expansion following co-culture with bone marrow-derived mesenchymal stem cells. Bone Marrow Transplant 2006; 37: 359- 66. [79] Walenda T, Bokermann G, Ventura Ferreira MS, et al. Synergistic effects of growth factors and mesenchymal stromal cells for expansion of hematopoietic stem and progenitor cells. Exp Hematol 2011; 39: 617-28. [80] Robinson SN, Simmons PJ, Yang H, Alousi AM, de Lima JM, Shpall EJ. Mesenchymal stem cells in ex vivo cord blood expansion. Best Pract Res Clin Haematol 2011; 24: 83-92. [81] Friedman R, Betancur M, Boissel L, Tuncer H, Cetrulo C, Klingemann H. Umbilical cord mesenchymal stem cells: adjuvants for human cell transplantation. Biol Blood Marrow Transplant 2007; 13: 1477-86. [82] Bakshi T, Zabriskie RC, Bodie S, et al. Mesenchymal stem cells 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.
  • 12. 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, Bongso A. Human umbilical cord Wharton's jelly stem cells and its conditioned medium support hematopoietic stem cell expansion ex vivo. J Cell Biochem 2012; 113: 658-68. [84] Bacigalupo A. Management of acute graft-versus-host disease. Br J Haematol 2007; 137: 87-98. [85] Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet 2009; 373: 1550-61. [86] Ringden O, Le Blanc K. Mesenchymal stem cells for treatment of acute and chronic graft-versus-host disease, tissue toxicity and hemorrhages. Best Pract Res Clin Haematol 2011; 24: 65-72. [87] Wu KH, Chan CK, Tsai C, et al. Effective treatment of severe steroid-resistant acute graft-versus-host disease with umbilical cord-derived mesenchymal stem cells. Transplantation 2011;91:1412-6. [88] Choi SW, Levine JE, Ferrara JL. Pathogenesis and management of graft-versus-host disease. Immunol Allergy Clin North Am 2010; 30: 75-101. [89] Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood 2008; 112: 4371-83. [90] Sprangers B, Fevery S, Van WB, De Somer L, Waer M, Billiau AD. Can graft-versus-leukemia reactivity be dissociated from graft- versus-host disease? Front Biosci 2007; 12: 4568-94. [91] Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002; 295: 2097-100. [92] Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 1999; 94(1): 333-9. [93] Boissel L, Tuncer HH, Betancur M, Wolfberg A, Klingemann H. Umbilical cord mesenchymal stem cells increase expansion of cord blood natural killer cells. Biol Blood Marrow Transplant 2008; 14: 1031-8. [94] Sun L, Akiyama K, Zhang H, et al. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells 2009; 27: 1421-32. [95] Sun L, Wang D, Liang J, et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum 2010; 62: 2467-75. [96] Shi D, Wang D, Li X, et al. Allogeneic transplantation of umbilical cord-derived mesenchymal stem cells for diffuse alveolar hemorrhage in systemic lupus erythematosus. Clin Rheumatol 2012; 31: 841-6. [97] Rachakatla RS, Pyle MM, Ayuzawa R, et al. Combination treatment of human umbilical cord matrix stem cell-based interferon-beta gene therapy and 5-fluorouracil significantly reduces growth of metastatic human breast cancer in SCID mouse lungs. Cancer Invest 2008; 26: 662-70. [98] Ayuzawa R, Doi C, Rachakatla RS, et al. Naive human umbilical cord matrix derived stem cells significantly attenuate growth of human breast cancer cells in vitro and in vivo. Cancer Lett 2009; 280: 31-7. [99] Gauthaman K, Fong CY, Arularasu S, et al. Human Wharton's jelly stem cell conditioned medium and cell-free lysate inhibit human osteosarcoma and mammary carcinoma cell growth in vitro and in xenograft mice. J Cell Biochem 2013; 114: 366-77. [100] Moretti P, Hatlapatka T, Marten D, et al. Mesenchymal stromal cells derived from human umbilical cord tissues: primitive cells with potential for clinical and tissue engineering applications. Adv Biochem Eng Biotechnol 2010; 123: 29-54. [101] Mariotti E, Mirabelli P, Abate G, et al. Comparative characteristics of mesenchymal stem cells from human bone marrow and placenta: CD10, CD49d, and CD56 make a difference. Stem Cells Dev 2008; 17: 1039-41. [102] Delorme B, Ringe J, Gallay N, et al. Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood 2008; 111: 2631-5. [103] Farias VA, Linares-Fernandez JL, Penalver JL, et al. Human umbilical cord stromal stem cell express CD10 and exert contractile properties. Placenta 2011; 32: 86-95 [104] Ding DC, Shyu WC, Chiang MF, et al. Enhancement of neuroplasticity through upregulation of beta1-integrin in human umbilical cord-derived stromal cell implanted stroke model. Neurobiol Dis 2007; 27: 339-53. [105] Prasanna SJ, Gopalakrishnan D, Shankar SR, Vasandan AB. Proinflammatory cytokines, IFNgamma and TNFalpha, influence immune properties of human bone marrow and Wharton jelly mesenchymal stem cells differentially. PLoS One 2010; 5: e9016. [106] Qiao C, Xu W, Zhu W, et al. Human mesenchymal stem cells isolated from the umbilical cord. Cell Biol Int 2008; 32: 8-15. [107] Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE. Human mesenchymal stem cells self-renew and differentiate according to a deterministic hierarchy. PLoS One 2009; 4: e6498. [108] Angelucci S,Marchisio M,Di Giuseppe F, et al. Proteome analysis of human Wharton's jelly cells during in vitro expansion. ProteomeSci 2010; 8: 18. [109] Gindraux F, Selmani Z, Obert L, et al. Human and rodent bone marrow mesenchymal stem cells that express primitive stem cells mark primitive stem cell markers can be directly enriched by using the CD49a molecule. Cell Tissue Res 2007; 327: 471-83. [110] Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 2003; 3: 890-6. [111] Selmani Z, Naji A, Zidi I, et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells 2008; 26: 212-22. [112] Struys T, Moreels M, Martens W, Donders R, Wolfs E, LambrichtsI. Ultrastructural and immunocytochemical analysis of multilineag differentiated human dental pulp- and umbilical cord- derived mesenchymal stem cells. Cells Tissues Organs 2011; 193: 366-78. [113] Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 1991; 78: 55-62. [114] Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 2007; 109: 1743-51. [115] Riekstina U, Cakstina I, Parfejevs V, et al. Embryonic stem cell marker expression pattern in human mesenchymal stem cells derived from bone marrow, adipose tissue, heart and dermis. Stem Cell Rev 2009; 5: 378-86. Received: October 16, 2012 Revised: December 17, 2012 Accepted: December 28, 2012