Vous êtes sur la page 1sur 19

Downloaded from rsif.royalsocietypublishing.

org on August 28, 2010

Biological characteristics of stem cells from foetal, cord blood


and extraembryonic tissues
Hassan Abdulrazzak, Dafni Moschidou, Gemma Jones and Pascale V. Guillot
J. R. Soc. Interface published online 25 August 2010
doi: 10.1098/rsif.2010.0347.focus

References This article cites 205 articles, 39 of which can be accessed free
http://rsif.royalsocietypublishing.org/content/early/2010/08/24/rsif.2010.0347.focus.full.
html#ref-list-1

P<P Published online 25 August 2010 in advance of the print journal.

Rapid response Respond to this article


http://rsif.royalsocietypublishing.org/letters/submit/royinterface;rsif.2010.0347.focusv1

Subject collections Articles on similar topics can be found in the following collections

bioengineering (85 articles)


biomedical engineering (188 articles)

Email alerting service Receive free email alerts when new articles cite this article - sign up in the box at the top
right-hand corner of the article or click here

Advance online articles have been peer reviewed and accepted for publication but have not yet appeared in
the paper journal (edited, typeset versions may be posted when available prior to final publication). Advance
online articles are citable and establish publication priority; they are indexed by PubMed from initial publication.
Citations to Advance online articles must include the digital object identifier (DOIs) and date of initial
publication.

To subscribe to J. R. Soc. Interface go to: http://rsif.royalsocietypublishing.org/subscriptions

This journal is © 2010 The Royal Society


Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

J. R. Soc. Interface
doi:10.1098/rsif.2010.0347.focus
Published online

REVIEW

Biological characteristics of stem cells


from foetal, cord blood and
extraembryonic tissues
Hassan Abdulrazzak, Dafni Moschidou, Gemma Jones
and Pascale V. Guillot*
Institute of Reproductive and Developmental Biology, Imperial College London,
London W12 0NN, UK
Foetal stem cells (FSCs) can be isolated during gestation from many different tissues such as
blood, liver and bone marrow as well as from a variety of extraembryonic tissues such as
amniotic fluid and placenta. Strong evidence suggests that these cells differ on many biolo-
gical aspects such as growth kinetics, morphology, immunophenotype, differentiation
potential and engraftment capacity in vivo. Despite these differences, FSCs appear to be
more primitive and have greater multi-potentiality than their adult counterparts. For
example, foetal blood haemopoietic stem cells proliferate more rapidly than those found in
cord blood or adult bone marrow. These features have led to FSCs being investigated for
pre- and post-natal cell therapy and regenerative medicine applications. The cells have
been used in pre-clinical studies to treat a wide range of diseases such as skeletal dysplasia,
diaphragmatic hernia and respiratory failure, white matter damage, renal pathologies as
well as cancers. Their intermediate state between adult and embryonic stem cells also
makes them an ideal candidate for reprogramming to the pluripotent status.

Keywords: stem cell; foetal; regenerative medicine; induced pluripotent stem

1. INTRODUCTION work has shown that ES cells can be derived from


single blastomeres isolated using procedures similar to
Stem cells are undifferentiated cells with the capacity to
those routinely used for pre-implantation genetic diag-
self-renew, differentiate and repopulate a host in vivo
nosis (Klimanskaya et al. 2007). However, safety
(Weissman et al. 2001). Their plasticity or potency is
concerns still remain because of the tumorigenicity of
hierarchical ranging from totipotent (differentiating
ES cells. Alternatively, adult stem cells can be found
into all cell types including placenta), pluripotent (dif-
in almost all tissues examined including brain, dental
ferentiating into cells of the three germ layers,
pulp, muscle, bone marrow, skin and pancreas and
ectoderm, mesoderm and endoderm, but not tropho-
have been extensively characterized for their thera-
blastic cells), multipotent (differentiating into cells of
peutic potential. The adult stem cell could be multi-
more than one type but not necessarily into all the
potent (e.g. haematopoietic stem cells (HSCs) giving
cells of a given germ layer) to unipotent (differentiating
rise to all blood cells and adherent stromal/mesenchy-
into one type of cell only, e.g. muscle or neuron).
mal stem cells (MSCs) that give rise to bone, fat,
Pluripotent stem cells can be derived from the inner
cartilage and muscle) or unipotent (e.g. progenitor
cell mass of the pre-implantation embryo (i.e. embryo-
cells). Adult MSCs have the problem of being difficult
nic stem (ES) cells) or isolated from the foetal
to extract in sufficient numbers for therapy and/or pre-
primordial germ cell pool (PGC) above the allantois
senting restricted plasticity and limited proliferative
(i.e. embryonic germ (EG) cells and embryonic carci-
capacity compared with ES cells.
noma (EC) cells; Thomson et al. 1998; Andrews et al.
In recent years, foetal stem cells (FSCs) and stem
2005). The destruction of the blastocyst or early
cells isolated from cord blood or extraembryonic tissues
foetus necessary for their derivation/isolation raises
have emerged as a potential ‘half way house’ between
ethical concerns (Lo & Parham 2009), although recent
ES cells and adult stem cells. FSCs can be found
in foetal tissues such as blood, liver, bone marrow,
*Author for correspondence ( pascale.guillot@imperial.ac.uk). pancreas, spleen and kidney, and stem cells are also
One contribution to a Theme Supplement ‘Translation and found in cord blood and extraembryonic tissues such
commercialization of regenerative medicines’. as amniotic fluid, placenta and amnion (Marcus &

Received 30 June 2010


Accepted 5 August 2010 1 This journal is # 2010 The Royal Society
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

2 Review. Characteristics of stem cells H. Abdulrazzak et al.

Woodbury 2008). Their primitive properties, expansion fibronectin (Guillot et al. 2006, 2007a). Contrary to
potential and lack of tumorogenicity make them an adult bone marrow MSCs, first-trimester foetal blood,
attractive option for regenerative medicine in cell liver and bone marrow MSCs express baseline levels of
therapy and tissue engineering settings. While extraem- the pluripotency stem cell markers Oct-4, Nanog, Rex-1,
bryonic tissues could be used with few ethical SSEA-3, SSEA-4, Tra-1-60 and Tra-1-81 (Guillot et al.
reservations, the isolation of FSCs from abortuses is 2007b; Zhang et al. 2009). Regardless of their tissue of
subject to significant public unease. We review here origin, first-trimester foetal MSCs self-renew faster in cul-
the phenotypic characteristics of stem cells from ture than adult MSCs (30–35 h versus 80–100 h) and
foetal, cord blood and extraembryonic tissues, their senesce later while retaining a stable phenotype (Guillot
application in cell therapy and their potential for repro- et al. 2007b). Foetal MSCs are therefore more readily
gramming towards pluripotentiality. expandable to therapeutic scales for either pre- or post-
natal ex vivo gene or cell therapy and tissue engineering.
Having greater multi-potentiality and differentiating
2. PHENOTYPIC CHARACTERISTICS OF more readily than adult MSCs into cells of mesodermal
FOETAL STEM CELLS origin such as bone and muscle, they can also differentiate
Stem cells are collected from abortal foetal tissue, into cells from other lineages such as oligodendrocytes
surplus of pre-natal diagnostic tissues or tissues at and haematopoietic cells (MacKenzie et al. 2001; Chan
delivery, subject to informed consent, institutional et al. 2006; Kennea et al. 2009). In terms of engraftment,
ethics approval and compliance with national guidelines they have a competitive advantage compared with adult
covering foetal tissue research. Until recently they cells (Rebel et al. 1996; Harrison et al. 1997; Taylor et al.
were thought to be multi-potent (O’Donoghue & Fisk 2002). Other advantageous characteristics relevant to cell
2004), but this picture is now changing as evidence is therapy include having active telomerase (Guillot et al.
mounting regarding the existence of pluripotent sub- 2007b), expressing low levels of HLA I and lacking intra-
populations in some foetal and extraembryonic tissues cellular HLA II, and taking longer to express this upon
(Cananzi et al. 2009). Some of the properties of the interferon g stimulation than adult MSCs (Gotherstrom
cells are summarized in table 1. et al. 2004). Foetal MSCs also express a shared a2, a4
and a5b1 integrin profile with first-trimester HSCs, impli-
2.1. Foetal tissues cating them in homing and engraftment, and, consistent
with this, they have significantly greater binding to
2.1.1. Foetal MSCs. MSCs are multi-potent stem cells their respective extracellular matrix ligands than adult
that can differentiate towards mesoderm-derived MSCs (de la Fuente et al. 2003). Finally, human foetal
lineages (i.e. osteogenic, adipogenic, chondrogenic and MSCs are readily transducible with transduction efficien-
myogenic; Pittenger et al. 1999). Their potential to cies of greater than 95 per cent using lentiviral vectors
repair damaged tissue and their immunomodulatory with stable gene expression at both short- and long-time
properties make them very attractive for a wide range points without affecting self-renewal or multi-potentiality
of regenerative medicine applications such as cell (Chan et al. 2005).
therapy and tissue engineering for hollow and solid
organs (Horwitz et al. 2002; Abdallah & Kassem
2008; Le Blanc et al. 2008). They were first identified 2.1.1.2. Specific characteristics. Although the above
in adult bone marrow where they represent 0.001– characteristics are shared among the various types of
0.01% of total nucleated cells (Owen & Friedenstein foetal stem, they also have specific traits that are depen-
1988). Adherent stromal cells with similar characteristics dent on tissue of origin and gestational age. For example,
were subsequently isolated from other tissues such as adi- foetal MSCs are present in foetal blood from the earliest
pose, dental pulp, muscle, liver and brain (Porada et al. gestation tested, seven weeks, where they account for
2006; Huang et al. 2009). They are also found in foetal approximately 0.4 per cent of foetal nucleated cells.
and extraembryonic tissues, where they seem to possess Their numbers decline to very low levels after 13
greater proliferation capacity and differentiation poten- weeks’ gestation (Campagnoli et al. 2001). MSC derived
tial than their adult counterparts (Campagnoli et al. from foetal bone marrow also decline with age with one
2001; Guillot et al. 2007b; Pappa & Anagnou 2009). MSC found among 10 000 in mid-trimester as compared
with one MSC per 250 000 cells in adult marrow.
2.1.1.1. Common characteristics. MSCs isolated from Significant variations have been found among foetal
foetal tissues such as blood, liver, bone marrow, lung tissue cells in terms of cell surface markers. For
and pancreas all share common characteristics. For example, foetal lung has a higher proportion of
example, they are spindle-shaped cells with the capacity CD34þ/CD452 cells (44%) than bone marrow (4.8%),
to differentiate into the standard mesenchymal spleen (12.6%) and liver (7.5%) (in’t Anker et al.
lineages, i.e. bone, fat and cartilage. They do not 2003a,b). Also foetal metanephric MSCs do not express
express haematopoietic or endothelial markers (e.g. CD45, CD34 or other haematopoietic markers; instead,
they are CD452/342/142 and von Willebrand factor they express high levels of mesenchymal markers such
negative; Gucciardo et al. 2009), but express stroma- as vimentin, laminin and type I collagen.
associated markers CD29 (b1-integrin), CD73 (SH3 There is also variation among the cells in terms of
and SH4), CD105 (SH2), CD44 (HCAM1), the early differentiation capacity. MSCs derived from foetal
bone marrow progenitor cell marker CD90 (thy-1) and liver during the first and second trimester have a
the extracellular matrix proteins vimentin, laminin and reduced osteogenic differentiation potential in

J. R. Soc. Interface
Table 1. Characteristics of stem cells. E, embryonic; MSC, mesenchymal; H, haematopoietic; EP, epithelial; VSEL, very small embryonic like; P, pluripotent; M, multipotent; U,
unipotent; n.d., not determined. Asterisk denotes that the characteristics of these cells are not listed in the table (Kyo et al. 1997; Campagnoli et al. 2001; Gammaitoni et al. 2004; Miki
et al. 2005, 2007; Pranke et al. 2005; Dan et al. 2006; Habich et al. 2006; Miki & Strom 2006; Portmann-Lanz et al. 2006; Zhao et al. 2006; De Coppi et al. 2007a; Fong et al. 2007; Guillot

J. R. Soc. Interface
et al. 2007b; Ilancheran et al. 2007; Karahuseyinoglu et al. 2007; Kim et al. 2007; Li et al. 2007; Musina et al. 2007; Prat-Vidal et al. 2007; Degistirici et al. 2008; Moon et al. 2008;
Mountford 2008; Poloni et al. 2008; Sessarego et al. 2008; Troyer & Weiss 2008; Brooke et al. 2009; Cananzi et al. 2009; Riekstina et al. 2009; Zhang et al. 2009; Alaminos et al. 2010;
Anzalone et al. 2010; Castrechini et al. 2010; Hsieh et al. 2010; Insausti et al. 2010; Vaziri et al. 2010).

foetal tissues extraembryonic

embryonic stem cord Wharton’s amnion-derived amnion- chorion-


cells blood liver bone marrow blood jelly amniotic fluid epithelial cells derived MSC derived MSC adult BM MSC

cell phenotype (s) E MSC/H* MSC/H* MSC/H* MSC/H* MSC MSC/AFS/VSEL* EP MSC MSC MSC
doubling time (DT)/ 48– 24 h (DT)/ 21.9 + 1.1 h 46 h (second- 32 h (second- 39– 43 h 85– 11 h 25–38 h (second- 38.4 h (DT) 30c (PD) 21.25 + 3.2 h 54–111 h (DT)/
population doublings indefinite (first-trimester trimester trimester (DT)/ (DT) trimester MSC) (DT)/30d 7.1a (PD)
(PD) (PD) MSC) (DT)/ MSC) (DT) MSC) 20b 36 h (AFS) (DT)/
28.4a (PD) (DT) (PD) 66 (MSC)/over
250 AFS (PD)
feeder/matrigel þ 2 2 2 2 2 2 2 2 2 2
requirement
potency P P/M P/M P/M P/M P/M P/M P/M P/M P/M M
Oct-4 þþþ þþ þþ þ þ þ þ þ þ þ +
Sox2 þþþ n.d. n.d. n.d. þ 2 þ n.d. n.d. +
Nanog þþþ þþ þþ þþ þ þ + þ n.d. þ +
c-Myc þþþ n.d. n.d. n.d. n.d. n.d. þ þ n.d. n.d. n.d.
Klf-4 þþþ n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d.
TRF length/telomerase þþþ þþ þþ þþ þþ þ n.d. 2 n.d. þ 2
activity
alkaline phosphatase þþþ n.d. n.d. þþ þ þ þ 2 n.d. +
c-Kit (CD117) þþþ þ þ þ + þ þ + + 2 2
Rex-1 þþþ þþ þ þþ þ n.d. þ þ 2 þ +
SSEA-4 þþþ þþ þþ þþ þ þ þ þ 2 þ 2
SSEA-3 þþþ þþ þþ þþ þ 2 2 þ(low) 2 þ 2
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Tra-1-81 þþþ þþ þþ þþ n.d. þ 2 þ 2 þ 2


Tra-1-61 þþþ þþ þþ þþ n.d. þ + þ 2 þ 2
Review. Characteristics of stem cells

MHC class I þ(low) þ(low) þ(low) þ(low) þ þ þ þ(low) þ(low) þ þ


MHC class II (HLA-DR) 2 2 2 2 + 2 + 2 2 2 2
teratoma formation in yes n.d. n.d. n.d. n.d. n.d. no no n.d. n.d. no
immune compromised
mice
a
Over a period of 50 days.
b
Over eight passages.
c
Over 14 passages.
d
Over five passages.
H. Abdulrazzak et al. 3
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

4 Review. Characteristics of stem cells H. Abdulrazzak et al.

comparison with first-trimester foetal blood and with blood, suggesting that these cells are more primitive
second-trimester spleen, lung and bone marrow MSCs, and have greater potential than HSCs circulating later
possibly because of a reduction in osteogenic progenitor in ontogeny (O’Donoghue & Fisk 2004). Even relatively
numbers (in‘t Anker et al. 2003a,b). Foetal pancreatic differentiated erythroblasts are more primitive in first
MSCs, shown to be capable of differentiating into chon- compared with later trimester foetal blood. Foetal
drogenic, osteogenic or adipogenic lineages (Hu et al. blood HSCs proliferate more rapidly than those in
2003), can also engraft, differentiate and secrete cord blood or adult bone marrow, and produce all
human insulin in a sheep model (Ersek et al. 2010). haematopoietic lineages (Campagnoli et al. 2001).
Moreover, foetal pancreatic ductal stem cells can Foetal liver is also a source of HSCs, and, despite
differentiate into insulin-producing cells in vitro (Yao their relatively small number, it is possible to generate
et al. 2004). sufficient numbers of cells for transplantation in four
Foetal metanephric MSCs can be induced in vitro to weeks (Rollini et al. 2004), although, to our knowledge,
adopt an osteogenic or myogenic phenotype and, when it has not been proved that foetal liver HSCs can suc-
transplanted in a foetal lamb model, they show long- cessfully transplant a human recipient. The phenotype
term persistence and site-specific differentiation of foetal liver HSCs changes with gestation age. For
(Almeida-Porada et al. 2002). example, their differentiation potential decreases mark-
Altogether these data indicate a high level of edly with gestational age, and cells that expressed both
heterogeneity within the foetal stem cell pool. CD34 and c-kit (CD117) were identified in the first-
trimester foetal liver whereas the cells expressing more
committed hepatic markers only appeared during the
2.1.2. Foetal HSCs. HSCs are multi-potent stem cells second trimester (Nava et al. 2005).
that maintain functional haematopoiesis by generation
of all haematopoietic lineages throughout foetal and
2.2. Extraembryonic tissue
adult life (Weissman & Shizuru 2008). They are charac-
terized by the expression of CD34 and CD45 antigens, 2.2.1. Umbilical cord blood. Umbilical cord blood is now
and the absence of markers such as CD38 and human an established source of transplantable HSCs that have
leucocyte antigen (HLA)/DRE (Huss 2000; Taylor a greater proliferative capacity, lower immunological
et al. 2002). During ontogeny the site of haematopoiesis reactivity and lower risk of graft-versus-host disease
is modified several times (Cumano et al. 2001). An area (GVHD) than those derived from adult bone marrow
along the dorsal embryonic aorta termed the aorta– (Broxmeyer et al. 1989; Yu et al. 2001; Ballen 2005;
gonad–mesonephros (AGM) is a rich source of HSCs, Schoemans et al. 2006; Brunstein et al. 2007; Hwang
which then migrate to the embryonic liver and later et al. 2007; Broxmeyer 2010). These cells are capable
seed other haematopoietic tissues such as the bone of repopulating bone following intra-bone injection of
marrow (McGrath & Palis 2008). HSCs are usually severe combined immunodeficiency mice (Mazurier
assayed in animal models based on their capacity to et al. 2003; Wang et al. 2003) and are used clinically
repopulate the entire haematopoietic system in con- as an alternative to adult bone marrow HSCs in some
ditioned recipients after transplantation. Similar to cases (Delaney et al. 2010). The cells are CD34þ and
MSCs, it is possible to transplant human cells into xeno- CD382 and their frequency is greater than that of
geneic recipients such as foetal sheep that act as models bone marrow or peripheral blood following cytokine
of human haematopoiesis (O’Donoghue & Fisk 2004). mobilization (Pappa & Anagnou 2009). It has also
First-trimester foetal blood contains more CD34þ been demonstrated that cord blood contains MSCs
cells than term gestation blood, yet CD34þ cells (Weiss & Troyer 2006; Secco et al. 2008) that can sup-
account for only 5 per cent of CD45þ cells in the port the in vivo expansion of HSCs and function as an
blood at that stage (Campagnoli et al. 2001). The accessory cell population for engraftment ( Javazon
number of circulating HSCs increases from the first tri- et al. 2004; Wang, J. et al. 2004). The frequency of
mester to peak in the second trimester in utero, MSCs in umbilical cord blood is low however, with
probably because of cells migrating from the foetal MSCs successfully isolated from only a third of all
liver to establish haematopoiesis in the foetal bone samples collected (Bieback et al. 2004).
marrow (Clapp et al. 1995). Some HSCs remain in the Cord blood stem cells expressing baseline levels of ES
umbilical cord at delivery, where they can be collected cell markers such as Oct-4, Nanog, SSEA-3 and SSEA-4
for allogeneic or occasionally autologous cell transplan- have also been described (Zhao et al. 2006). Using a
tation (see §2.2.1.). two-stage isolation approach whereby the erythrocytes
In the second trimester, CD34þ cells constitute 4 per in cord blood are lysed and the remaining cells are
cent of cells in blood, 16.5 per cent in bone marrow, sorted using flow cytometry, it has been possible to iso-
6 per cent in liver, 5 per cent in spleen and 1.1 per late and enrich for a subpopulation of cells that are
cent in the thymus (Lim et al. 2005). This frequency CXCR4þ, CD133þ, CD34þ, Lin2 and CD452.
of CD34þ cells in the blood gradually diminishes These cells have been described as very small
during the third trimester, probably reflecting establish- embryonic-like (VSEL) stem cells because they are
ment of the marrow as the primary site of only 3 – 5 mm in diameter and express Oct-4, Nanog
haematopoiesis and the declining role of the foetal and SSEA-4 (Kucia et al. 2007; Zuba-Surma et al.
liver in that regard ( Jones et al. 1994; Wagers et al. 2010). Compared with MSCs, VSEL cells are smaller,
2002). The number of cells negative for CD38 within have a large nucleus to cytoplasm ratio and open-type
the CD34þ population is also higher in early foetal chromatin. As a similar population has also been

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 5

found in adult bone marrow, where they also express the De Coppi et al. isolated c-kit-positive (CD117) cells
same primitive markers, it has been hypothesized that that represent about 1 per cent of cells present in
VSEL cells are related to a population of early PGCs second-trimester amniotic fluid. These cells were
that are retained during development (Kucia et al. named amniotic fluid stem (AFS) cells. They can be
2006). cultured without feeders, double in 36 h, are not
tumorigenic, have long telomeres and retain a normal
karyotype for over 250 population doublings (De
2.2.2. Wharton’s jelly. Wharton’s jelly is the connective
Coppi et al. 2007a). Cultured human AFS cells are posi-
tissue surrounding the umbilical vessels and includes
tive for ES cell (e.g. Oct-4, Nanog and SSEA-4) and
the perivascular, intervascular and subamnion regions
mesenchymal cell markers such as CD90, CD105
(Troyer & Weiss 2008). MSCs have been isolated from
(SH2), CD73 (SH3/4) and several adhesion molecules
all three regions and it remains unclear whether they
(e.g. CD29 and CD44; Tsai et al. 2006; Chambers et al.
represent distinct cell populations (Karahuseyinoglu
2007; De Coppi et al. 2007a). Furthermore, it was poss-
et al. 2007). The cells have been given different names
ible to generate clonal human lines from these cells,
by different groups (Troyer & Weiss 2008). MSCs iso-
verified by retroviral marking, which were capable of dif-
lated from Wharton’s jelly share similar properties
ferentiating into lineages representative of all three EG
with other cord blood MSCs as well as adult bone
layers. Almost all clonal AFS cell lines express Oct-4
marrow MSCs on the expression of markers, differen-
and Nanog, markers of a pluripotent undifferentiated
tiation potential and cytokine production (McElreavey
state (De Coppi et al. 2007a). c-kitþ Lin2 cells from
et al. 1991; Wang, H.-S. et al. 2004; Anzalone et al.
human and mouse amniotic fluid display a multi-lineage
2010). Wharton’s jelly MSCs can also be induced to
haematopoietic potential in vitro and in vivo, despite
differentiate into cells similar to neural, expressing
having low or negative CD34 expression (Ditadi et al.
neuron-specific enolase and other neural markers
2009). c-kit-positive human AFS cell lines can also
(Mitchell et al. 2003; Anzalone et al. 2010).
form embryoid bodies (EB) with an incidence of
18–82%. EB formation was accompanied by a decrease
2.2.3. Amniotic fluid. In recent years, amniotic fluid has in Oct-4 and Nodal and the induction of differentiation
emerged as a major source of putative pluripotent stem markers such as Pax 6, Nestin (ectodermal), GATA 4
cells that avoid many of the problems associated with and HBE1 (mesodermal) (Valli et al. 2010). The poten-
ES cells such as their non-suitability for autologous tial for a wide range of regenerative medicine and tissue
use, their capacity for tumour formation and the ethical engineering applications as well as their relatively few
concerns they raise. ethical concerns makes them an attractive source
In humans, the amniotic fluid starts to appear at the for cell therapy, particularly considering that a bank of
beginning of week 2 of gestation as a small film of 100 000 amniotic fluid specimens could potentially
liquid between the cells of the epiblast. The fluid expands supply 99 per cent of the US population with a perfect
separating the epiblast (i.e. the future embryo) from the match for transplantation (Atala 2009).
amnioblasts (i.e. the future amnion), thus forming the
amniotic cavity (Miki & Strom 2006). The origin of
amniotic fluid cells is still very much debatable 2.2.4. Placenta. The placenta is a fetomaternal organ
(Kunisaki et al. 2007; Cananzi et al. 2009). However, involved in maintaining foetal tolerance and allows
what is known is that the majority of cells present are nutrient uptake and gas exchange with the mother,
terminally differentiated and have limited proliferative but also contains a high number of progenitor cells or
capacity (Gosden & Brock 1978; Siegel et al. 2007). How- stem cells (Parolini et al. 2010). It has two sides: one
ever, a number of studies have demonstrated the presence foetal (amnion and chorion) and one maternal (decid-
of a subset of cells with a proliferative and differentiation uas). The amnion membrane contains two cell types:
potential (Torricelli et al. 1993; Streubel et al. 1996). the amniotic epithelial cells (AECs) derived from the
A variety of different types of stem cells have been iso- epiblast; and amniotic mesenchymal cells derived from
lated and characterized from amniotic fluid. These the hypoblast. The chorion layer consists of cytotropho-
include cells found in mid-gestation expressing the haema- blasts and syncytiotrophoblasts that are derived from
topoietic marker CD34 (Da Sacco et al. 2010) as well as the outer layer of the blastocyst (trophectoderm)
cells with mesenchymal features, able to proliferate in (Ilancheran et al. 2009; Insausti et al. 2010).
vitro more rapidly than comparable foetal and adult The availability, phenotypic plasticity and immuno-
cells (Kaviani et al. 2001; in‘t Anker et al. 2003a,b; modulatory properties of placenta-derived progenitor/
Nadri & Soleimani 2007; Roubelakis et al. 2007; Sessarego stem cells are useful characteristics for cell therapy
et al. 2008). Amniotic fluid MSCs are negative for and tissue engineering. Cells can be isolated during
haematopoietic markers such as CD45, CD34 and CD14 ongoing pregnancy using minimally invasive techniques
(Prusa & Hengstschlager 2002; in‘t Anker et al. 2003a,b; such as chorionic villus sampling (CVS) and placental
Prusa et al. 2003; Tsai et al. 2004). Despite their high pro- tissues are readily available at delivery for allogeneic
liferation rate, these cells display a normal karyotype or autologous use. Cells that have been isolated from
when expanded in vitro and do not form tumours in placenta include the human AECs, human amnion
vivo (Sessarego et al. 2008). They exhibit a broad differen- mesenchymal stromal stem cells (AMSCs), human
tiation potential towards mesenchymal lineages (in‘t chorionic mesenchymal/stromal stem cells (CMSCs),
Anker et al. 2003a,b; Kolambkar et al. 2007; Nadri & human chorionic trophoblastic cells and HSCs (Parolini
Soleimani 2007; Tsai et al. 2007). et al. 2008).

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

6 Review. Characteristics of stem cells H. Abdulrazzak et al.

Mesenchymal placenta cells are plastic adherent, term placenta-derived stem cells are found at the
share a similar immunophenotype and have lineage quiescent G0/G1 phase of the cell cycle.
differentiation potential. They express stromal markers
such as CD166, CD105, CD73, CD90 and others, while
they are negative for the haematopoietic markers CD14, 2.2.4.3. Chorion-derived stem cells. CMSCs have been
CD34 and CD45 (Igura et al. 2004; Sudo et al. 2007). isolated during pregnancy (CVS) or from the term pla-
Additionally, they express pluripotency markers such as centa at delivery. The immunophenotype of term
SSEA3, SSEA4, Oct-4, Nanog, Tra-1-60 and Tra-1-81 placenta cells is similar to that of adult bone marrow
(Yen et al. 2005; Battula et al. 2007). They lack or have MSCs, although renin and flt-1 have been shown to
very low expression of MHC class I antigens and do not be expressed uniquely in placenta MSCs (Fukuchi
express MHC class II antigens or the T-cell co-stimulatory et al. 2004). Similarly to human amnion isolated from
molecule B7, giving the cells immunomodulatory term placenta, term chorion MSCs successfully engraft
properties (Bailo et al. 2004; Miao et al. 2006). in neonatal swine and rats and failed to induce a xeno-
geneic response (Bailo et al. 2004), indicating that these
cells may have an immunoprivileged status consistent
2.2.4.1. Amniotic epithelial cells. AECs are commonly with their low level of HLA I and absence of HLA II
isolated from the amniotic membrane using digestive expression (Kubo et al. 2001). Of relevance for pre-
enzymes. The cells are plastic adherent and grow under natal autologous and allogeneic therapy, chorion
MSC conditions, i.e. Dulbecco’s modified Eagle’s MSCs can be successfully isolated from chorionic villi
medium (DMEM) supplemented with 10–20% FBS, during first-trimester gestation. The chorionic villi do
with the addition of growth factors such as epidermal not express typical MSC cell surface antigens and
growth factor (EGF) (Miki et al. 2010). c-Kit and CD90 have the capacity to differentiate into lineages of the
(Thy-1) expression is either negative or at a low level three germ layers, with a subset of cells expressing the
(Miki et al. 2005; Miki & Strom 2006). However, evidence pluripotency markers Oct-4, ALP, Nanog and Sox2
suggests that they express pluripotency markers and have (Spitalieri et al. 2009). In addition to differentiating
the ability to form xenogeneic chimera with mouse ES into adipogenic, chondrogenic and osteogenic cells in
cells in vitro (Tamagawa et al. 2004). The cells have sub- vitro (Portmann-Lanz et al. 2006; Ilancheran et al.
sequently been differentiated in vitro into cell types from 2007), they can also differentiate into cells with some
all three germ layers (Miki et al. 2005; Miki & Strom characteristics of hepatocytes in vitro and have the abil-
2006; Ilancheran et al. 2007; Parolini et al. 2008). ity to store glycogen (Chien et al. 2006; Huang 2007;
However, in contrast with ES cells, Miki et al. (2005) Tamagawa et al. 2007). Differentiation into other cell
demonstrated that human AE cells did not form gross types such as endothelial cells (Alviano et al. 2007),
or histological tumours up to seven months post- cardiomyocytes (Zhao et al. 2005), neurons, oligoden-
transplantation in SCID/Beige mice. drocytes and glial cells (Miao et al. 2006; Yen et al.
2008; Portmann-Lanz et al. 2010) has also been
reported, but these findings have mostly relied on upre-
2.2.4.2. Amnion-derived mesenchymal stromal cells. gulation of tissue-specific markers without providing
Both amnion and chorion MSCs have been extensively robust functionality tests.
characterized and can be isolated throughout gestation
from first trimester to delivery. Both cells share
common characteristics, such as plastic adherence,
3. THERAPUTIC APPLICATIONS OF
and, despite limited proliferation capacity, show in
FOETAL STEM CELLS
vitro differentiation down the osteogenic, adipogenic,
chrondrogenic and neurogenic lineages, although some Regenerative medicine aims to restore diseased or
report and show in vivo multi-organ engraftment damaged tissue using the body’s own cells in order to
capacity. In addition, the volume of term placenta overcome the shortage of donated organs and the risk
makes it an attractive source of stem cells, as on average associated with their rejection (Atala 2009). The use
human term placenta weighs more than 590 g of stem cells from foetal, cord blood and extraembryonic
(Bolisetty et al. 2002). Human placenta contains var- tissues for regenerative medicine applications has
ious types of cells of different developmental origin. increased over the past few years. However, the surpris-
Cells of the chorion and decidua are derived from tro- ing picture that has emerged is that cell replacement is
phectoderm, while amnion is derived from the epiblast probably not the major mechanism by which cell
of the developing embryo (Crane & Cheung 1988). therapy confers functional benefit. Rather, stem cells
MSCs have been successfully isolated from first-, act beneficially by exerting trophic effects on host tis-
second- and third-trimester placental compartments, sues. Moreover, because they can be expanded to very
including the amnion, chorion, decidua parietalis and large numbers compared with adult stem cells, they
decidua basalis (in‘t Anker et al. 2004; Portmann- are ideal candidates for seeding scaffolds and matrices
Lanz et al. 2006; Soncini et al. 2007; Poloni et al. used for tissue engineering applications such as the
2008), and represent less than 1 per cent of cells present repair of hollow and solid organs. In this section, we
in the human placenta (Zhang et al. 2004; Alviano et al. will consider some of the therapeutic applications
2007). First-trimester placenta stem cells generally grow FSCs have been put to in pre-clinical studies for repair-
faster than those found in the third trimester ing diseased or damaged tissue as well as in cancer
(Portmann-Lanz et al. 2006), and the majority of treatment (summarized examples are listed in table 2).

J. R. Soc. Interface
Table 2. Examples of therapeutic applications of foetal stem cells.

regenerated
cell type cell origin treated recipient tissue method of cell delivery disease treated outcome references

J. R. Soc. Interface
foetal tissues
blood and bone human oim mice bone intrauterine osteogenesis imperfecta transplantation resulted in decrease in Guillot et al. (2008a)
marrow transplantation (OI) fractures, increased bone strength, and unpublished
thickness and length. Transplanted data
cells were found clustered in areas of
active bone formation
human postnatal scid/ muscle intra-arterial delivery into Duchenne muscular galectin-1-exposed foetal MSCs formed Chan et al. (2006,
mdx dystrophic post-natal mice/ dystrophy (DMD) fourfold more human muscle fibres in 2007)
mouse model/ intrauterine post-natal scid/mdx mice than non-
prenatal Mdx transplantation using stimulated cells. In pre-natal
dystrophic mice intramuscular, immunocompetant mdx mice,
intravascular and intravascular delivery led to demise of
intraperitoneal delivery mice, intraperitoneal delivery led to
of cells into pre-natal systemic spread of cells and
mice intramuscular injections resulted in
local engraftment. The cells
differentiated at low level into
skeletal and myocardial muscle but
not at a curative level
liver human human foetus bone intrauterine OI cell engraftment of bone despite the Le Blanc et al. (2005)
diagnosed with transplantation donor cells being HLA-mismatched
severe OI with recipient. Bone histology showed
regularly arranged and configured
bone trabeculae
pancreas human sheep pancreas intrauterine type II diabetes the cells functionally engrafted in the Ersek et al. (2010)
transplantation pancreas in 50% of sheep. The
engrafted cells differentiated and were
able to secrete insulin
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

extraembryonic
Review. Characteristics of stem cells

cord blood human human bone systematic administration malignant and non- used in the treatment of leukaemia/ Broxmeyer (2010)
marrow malignant blood cancer, Fanconi anaemia, severe
disorders aplastic anaemia, myelodysplastic
syndrome and other disorders
human mouse pancreas intra peritoneal type II diabetes cord blood stem cells introduced into Zhao et al. (2006)
streptozotocin-induced diabetic
mouse model differentiated into
functional insulin-producing cells and
eliminated hyperglycaemia
(Continued.)
H. Abdulrazzak et al. 7
Table 2. (Continued.)

regenerated

J. R. Soc. Interface
cell type cell origin treated recipient tissue method of cell delivery disease treated outcome references

Wharton’s human rat brain injection into the brain Parkinson’s disease the cells ameliorated apomorphine- Weiss et al. (2006)
Jelly striatum induced rotations in a rat model of
Parkinson’s disease
amniotic fluid sheep sheep muscle cells seeded on collagen partial diaphragmatic diaphragmatic hernia recurrence was Fuchs et al. (2004)
hydrogel and placed on replacement significantly higher in animals that
intestinal cellular graft did not receive the cell grafts
rat rat smooth cells injected into site of wound healing of cells prevented cryo-injury-induced De Coppi et al.
muscle injury injured bladder hypertrophy of surviving bladder (2007a)
8 Review. Characteristics of stem cells

smooth muscle cells but failed to


differentiate specifically to smooth
muscle
mouse mouse brain intra ventricular injection focal cerebral the cells significantly reversed Rehni et al. (2007)
ischaemia neurological deficits in the treated
reperfusion injury animals
human mouse lung systematic administration lung injury AFS cells exhibited a strong tissue Carraro et al. (2008)
engraftment in a mouse lung injury
model and expressed specific alveolar
and bronchiolar epithelial markers
H. Abdulrazzak et al.

human rat heart intramyocardial injection myocardial infarction treated animals showed a preservation Yeh et al. (2010)
of the infarcted thickness,
attenuation of left ventricle
remodelling, a higher vascular density
and general improvement of cardiac
function
placenta human amnion mouse brain intracranial injection Parkinson’s disease cells transplanted into a mouse model of Kong et al. (2008)
AEC Parkinson’s disease differentiated into
neuron, astrocyte and
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

oligodendrocyte and promoted


endogenous neurogenesis
human amnion rat heart intramyocardial injection myocardial infarction cells integrated into normal and Zhao et al. (2005),
MSC infarcted rat cardiac tissue where Ventura et al.
they differentiated into (2007)
cardiomyocyte-like cells and resulted
in considerable improvements to
ventricular function, capillary density
and scar tissue
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 9

3.1. Bone Human AFS cells, transplanted into an immune-


competent and ciclosporin immune-suppressed rat
Osteogenesis imperfecta (OI) is characterized by osteo-
model of myocardial infarction, were rejected probably
penia and bone fragility due to abnormal collagen
because of the expression of B7 co-stimulatory
production caused by mutations in the a chains of col-
molecules as well as macrophage marker CD68.
lagen type I. First-trimester allogeneic HLA-
Furthermore, chondro-osteogenic cell masses were
mismatched male foetal liver MSCs were transplanted
observed in the infarcted hearts of some of the trans-
in utero into a female foetus diagnosed with severe
planted animals (Chiavegato et al. 2007). However, in
OI. Bone biopsy showed regularly arranged and config-
another study it was found that these masses occurred
ured bone trabeculae and no adverse immune reaction
independently of AFS cell injection and that the cells
was observed (Le Blanc et al. 2005). Using a mouse
did not increase the presence of these masses (Delo
model of OI, our group has shown that in utero trans-
et al. 2010).
plantation of foetal blood MSCs ameliorated the
Human AMSCs are able not only to express cardiac-
disease phenotype, producing a clinically relevant
specific genes under specific culture conditions, but also
two-thirds reduction in fracture incidence along with
to integrate into normal and infarcted rat cardiac tissue
an improvement in bone structure and mechanical
where they differentiate into cardiomyocyte-like cells
properties (Guillot et al. 2008a). Foetal bone
(Zhao et al. 2005). The cells result in considerable
marrow MSCs loaded on a highly porous scaffold were
improvements to ventricular function, capillary density
able to reach confluence within the scaffold more
and scar tissue (Ventura et al. 2007) and similar find-
quickly than umbilical cord and adult MSCs. Further-
ings have been reported using native rat AMSCs
more, they demonstrated higher in vitro and in vivo
(Fujimoto et al. 2009). Stem cells derived from pre-
osteogenic differentiation capacity, highlighting their
natal chorionic villi were also successfully engineered
suitability for bone tissue engineering applications
into a living autologous heart valve, which could have
(Zhang et al. 2009).
postnatal applications for repairing congenital cardiac
malformations (Schmidt et al. 2006).
3.2. Muscle
Duchenne muscular dystrophy (DMD) is an X-linked 3.3. Kidney
myopathy affecting one in 3500 boys. The main genetic
In a mouse model of collagen deficiency characterized
defect leads to absence of dystrophin, resulting in
by abnormal collagen deposition in renal glomeruli
muscle damage and wasting. The affected boys
(Phillips et al. 2002; Brodeur et al. 2007), we recently
become wheelchair-bound by 12 years of age and suc-
showed that intrauterine transplantation of foetal
cumb to respiratory failure or cardiopmyopathy by
blood MSCs led to a reduction of abnormal homotri-
the third or forth decade of life (Manzur & Muntoni
meric collagen type I deposition in the glomeruli of
2009). The main animal model for studying DMD is
4 – 12 week old col1a2-deficient mice. Furthermore, we
the mdx mouse that has a premature stop codon result-
showed that the damaged kidneys preferentially
ing in a termination in exon 23 of the dystrophin gene
recruited donor cells in the glomeruli. The study sup-
(Grounds et al. 2008). Galectin-1-treated foetal blood
ports the feasibility of pre-natal treatment for renal
and bone marrow MSCs, transplanted into postnatal
diseases such as Alport syndrome and the polycystic
severe combined immunodeficiency/dystrophic mice
kidney diseases (Guillot et al. 2008b).
(scid/mdx) as well immunodeficient mice whose
Acute tubular necrosis could also be potentially trea-
muscle was induced to regenerate by cryodamage,
ted with foetal cells. Human AFS cells injected into an
formed fourfold more human muscle fibres than non-
immunodeficient mouse model of the disease decreased
stimulated MSCs (Chan et al. 2006). Intrauterine
the number of damaged tubules and reduced apoptosis.
transplantation of foetal MSCs into immunocompetent
The cells also promoted the proliferation of tubular epi-
E14-16 mdx mice resulted in widespread long-term
thelial cells and appeared to have a beneficial
engraftment in multiple organs with a predilection for
immunomodulatory effect (Perin et al. 2010).
muscle compared with non-muscle tissues. However,
the engraftment level observed (0.5 – 1%) falls signifi-
cantly below the levels required for functional
3.4. Lung
improvement in DMD. The low engraftment might be
linked to an absence of muscle pathology at the time Human AFS cells injected into the tail vein of nude
of transplantation (Chan et al. 2007). mice following hyperoxia injury show the capacity to
AFS cells have been used in an ovine model of home to the lung and engraft. They also expressed
diaphragmatic hernia: repair of the muscle deficit specific alveolar and bronchiolar epithelial markers
using grafts engineered with autologous mesenchymal (e.g. TFF1, SPC and CC10; Carraro et al. 2008).
amniocytes leads to better structural and functional AECs and AMSCs from either human or mouse have
results than equivalent foetal myoblast-based and acel- been used in a mouse model of bleomycin-induced lung
lular grafts (Fuchs et al. 2004; Kunisaki et al. 2006). In injury and shown to cause a reduction in severity of
a rat model of bladder cryo-injury, AFS cells also show lung fibrosis. This occurs regardless of cell source (allo-
the ability to differentiate into smooth muscle and to geneic or xenogeneic) or administration route (systemic,
prevent the compensatory hypertrophy of surviving intravenous or intraperitoneal; local, intratracheal;
smooth muscle cells (De Coppi et al. 2007b). Cargnoni et al. 2009).

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

10 Review. Characteristics of stem cells H. Abdulrazzak et al.

3.5. Liver factor and fibroblast growth factor 2 (Lund et al.


2007). Interestingly, Wharton’s jelly MSCs trans-
Second-trimester foetal liver cells, enriched for CD326
planted into the vitreous demonstrated a rescue effect,
(hepatocyte progenitor marker) and labelled with Tc-
indicating that they could enhance the survival of
d,l-hexamethyl-propylene-amine oxime (Tc-HMPAO)
photoreceptor cells without being in close proximity
were infused into the hepatic artery of 25 patients
to them. This effect was presumably the result of
with end stage liver cirrhosis. The cells restored the
diffusible growth factors.
lost liver function in the patients without provoking
Very similar findings have also been reported for
an immune reaction, probably because of the absence
AECs that confer neuroprotection and functional recov-
of HLA class II expression of the infused cells (Khan
ery in animal models of Parkinson’s disease (Bankiewicz
et al. 2010).
et al. 1994; Kakishita et al. 2000, 2003; Kong et al.
Human AECs can secrete albumin in culture and,
2008) and ischaemia (Liu et al. 2008). The observed
when b-galactosidase-tagged AECs were transplanted
therapeutic effects are probably mediated by secretion
into immunodeficient mice, the cells integrated into
of diffusible factors, including neurotransmitters
the liver parenchyma and could be detected until day
(Elwan & Sakuragawa 1997; Sakuragawa et al. 1997,
7 post transplant (duration of study; Sakuragawa
2001) and neurotrophic and growth factors (Koizumi
et al. 2000). In another study, the engrafted cells
et al. 2000; Uchida et al. 2000).
resulted in the detection of human a-1 antitrypsin in
AFS cells can harbour trophic and protective effects
the serum of recipient animals, confirming that AECs
in the central and the peripheral nervous systems. Pan
are capable of performing this important hepatic
et al. (2006, 2007, 2009) showed that AFS cells facilitate
function in vivo (Miki & Strom 2006).
peripheral nerve regeneration after injury, particularly
if accompanied with suppressors of inflammatory cyto-
kines such as fermented soya bean extracts. After
3.6. Brain
transplantation into the striatum, AFS cells are capable
Human Wharton’s jelly MSCs ameliorate apomorphine- of surviving and integrating in the rat adult brain and
induced behavioural deficits in a hemiparkinsonian rat can migrate towards areas of ischaemic damage
model (Weiss et al. 2006). There was a significant (Cipriani et al. 2007). Moreover, the intra-ventricular
decrease in apomorphine-induced rotations at four administration of AFS cells in mice with focal cerebral
weeks continuing up to 12 weeks post transplantation ischaemia – reperfusion injuries significantly reverses
in Parkinson’s disease (PD) rats that received human neurological deficits in the treated animals (Rehni
Wharton’s jelly MSCs transplants compared with the et al. 2007).
PD rats that received a sham transplant. The behav-
ioural findings correlated with the numbers of tyrosine
3.7. Cancer therapy
hydroxylase-positive cell bodies observed in the mid-
brain following sacrifice at 12 weeks, indicating a HSCs from bone marrow or umbilical cord blood have
‘rescue from a distance’ phenomenon. One explanation long been used to re-establish the haematopoietic
for this effect may be that Wharton’s jelly MSCs system following radiation and/or chemotherapy
synthesize glial cell-derived neurotrophic factor (Osawa et al. 1996; Bhatia et al. 1997). More recently,
(GDNF), a potent survival factor for dopaminergic research has shown that MSCs are attracted to tumours
neurons, as well as other trophic factors such as because the latter often act as unresolved wounds pro-
vascular endothelial growth factor (VEGF) and ciliary ducing a continuous source of chemoattractant
neurotrophic factor. In another report, Wharton’s inflammatory mediators. This property is being
jelly MSCs were first induced towards dopaminergic exploited by using exogenously delivered MSCs as
neurons using neuron-conditioned media, sonic hedge- vehicles for delivering anti-cancer molecules to tumours
hog and fibroblast growth factor 8, and then (Loebinger & Janes 2010). Wharton’s jelly cells, like
transplanted into hemiparkinsonian rats (Fu et al. neural stem cells and MSCs (Aboody et al. 2000;
2006). Despite the fact that the rats were not immune Studeny et al. 2004), appear to migrate to areas of
suppressed, Wharton’s jelly MSCs were identified five tumour growth. Human breast carcinoma cells were
months later and prevented the progressive degener- intravenously injected into SCID mice, followed by
ation/behavioural deterioration seen in control rats intravenous transplantation of fluorescently labelled
with unilateral lesions. Similarly, rat Wharton’s jelly Wharton’s jelly MSCs. One week after transplant
cells transplanted into the brains of rats with global cer- Wharton’s jelly MSCs were found near or within lung
ebral ischaemia caused by cardiac arrest and tumours and not in other tissues. They were engineered
resuscitation significantly reduced neuronal loss, appar- to express human interferon beta and were administered
ently owing to a rescue phenomenon (Jomura et al. intravenously into SCID mice bearing tumours. This
2007). Lund et al. administered Wharton’s jelly MSCs treatment significantly reduced the tumour burden
into the eyes of a rodent model of retinal disease. (Rachakatla et al. 2007). Similarly, human umbilical
Here, Wharton’s jelly MSCs were compared with bone cord blood-derived MSCs (UCB – MSCs) have been
marrow-derived mesenchymal stem cells (BMSCs) and used to deliver a secretable trimeric form of tumour
placental stem cells. They reported that the Wharton’s necrosis factor-related apoptosis-inducing ligand
jelly MSCs exhibited the best histological evidence of (stTRAIL), via adenoviral transduction mediated by
photoreceptor rescue with increased production of cell-permeable peptides, to human glioma cells in
trophic factors such as brain-derived neurotrophic nude mice. The genetically modified cells showed

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 11

greater efficacy in terms of inhibiting tumour growth candidates for reprogramming and found that the iPS
and prolonging the survival of glioma-bearing mice cell colonies were generated twice as fast, yielding
compared with direct injection of adenovirus encoding nearly a 200 per cent increase in number compared
the stTRAIL gene into the tumour mass (Kim et al. with cultured adult skin cells. However, they did not
2008). provide absolute efficiency data (Galende et al. 2009).
Li et al. reprogrammed human amniotic fluid-derived
cells (hAFDCs) with efficiencies varying from 0.059 to
1.525 per cent. The iPS colonies generated expressed
4. USE OF FSCs FOR REPROGRAMMING
pluripotency markers such as Oct-4, Sox2 and SSEA 4
The major advantage of human ES cells over other stem and they maintained a normal karyotype (Li et al.
cell types is their capacity to differentiate into lineages 2009). Wharton’s jelly cells and term placenta cells
from the three germ layers. However, there are two have also been used, giving reprogramming efficiencies
major objections to their use. The moral objection is of 0.4 per cent and 0.1 per cent, respectively, which com-
that their derivation requires the destruction of pare rather well with efficiencies reported for adult
embryos. The practical objection is that they have a fibroblasts (less than 0.01%). The generated iPS cell
limited clinical application as they can only be used in colonies expressed several pluripotency markers, formed
an allogeneic fashion. This is also true for some foetal EB and, when injected into nude mice, they generated
tissues such as liver and bone marrow. However, extra- teratoma-containing derivates of the three germ layers
embryonic tissues can be used autologously. (Cai et al. 2010). Giorgetti et al. selected CD133þ (hae-
Thus, it was seen as advantageous to develop a matopoietic stem and progenitor cell marker) cord blood
method of creating ES-like cells from somatic cells. cells and managed to reprogramme them by retroviral
Early attempts at achieving this were nuclear trans- transduction with Oct-4 and Sox2 only without needing
plantation and fusion of somatic cells and ES cells additional chemical compounds. iPS cell colonies derived
( Jaenisch 2009). However, a considerable step forward from CD133þ cord blood cells (expressing pluripotency
was the generation of the induced pluripotent stem markers and capable of forming EB and teratomas)
(iPS) cells. The production of iPS cells with quasi- were generated in two weeks whereas no colonies were
identical genetic and functional properties offers the obtained with control keratinocytes or fibroblasts using
possibility to bypass both moral conflicts and different the two factors only (Giorgetti et al. 2009, 2010).
genetic background inherent to the technologies CD133þ cord blood cells and foetal NSCs already
mentioned above. iPS are developed from a non- have a baseline level expression of Oct-4 and/or Sox2
pluripotent cell, usually an adult somatic cell, by and that, along with a generally more permissive chro-
causing a forced expression of several genetic sequences matin organization, might be the key to their greater
and were first produced in 2006 by Takahashi & reprogramming efficiency compared with adult cells.
Yamanaka from mouse somatic cells. The key genes
Oct-4, the transcription factor Sox2, c-Myc protoonco-
gene protein and Klf4 (Krueppel-like factor 4) were 5. CONCLUSION
sufficient to reprogramme fibroblasts to cells closely Human FSCs can be isolated from foetal organs or
resembling ES cells (Takahashi & Yamanaka 2006; extraembryonic sources. They have the advantage of
Takahashi et al. 2007). rapid proliferation, stable karyotype and low or negli-
Despite the high similarity between iPS and ES cells, gible immunogenicity. Unlike ES cells, they do not
tumour formation in iPS cell chimeric mice was high, form teratomas in vivo and have far fewer ethical con-
presumably because of the expression of c-Myc in iPS cerns as they are mostly obtained from tissues that
cell-derived somatic cells (Maherali et al. 2007). would otherwise be discarded. Many of these cells
Thomson et al. showed that c-Myc was not necessary seem to express some of the same pluipotency markers
as they were able to generate iPS cells using Oct-4, found in ES cells, a feature largely absent from most
Sox2, Nanog and Lin28 using a lentiviral system (Yu adult-derived stem cells. They also have the further
et al. 2007). More recently, it was shown that human advantage over adult stem cells of senescing much
fibroblasts cells can be used to generate iPS cells if later and being more readily amenable to genetic
transduced with Oct-4 and Sox2 only in the presence modification.
of valproic acid (Huangfu et al. 2008). All of these features make them valuable for poten-
Terminally differentiated cells might not be the ideal tial therapy applications. Thus far they have been
candidate for iPS cell generation because a greater used in pre-clinical settings to treat a variety of diseases
number of steps could be required to ‘reprogramme’ such as osteogenesis imperfecta, congenital diaphrag-
their genome than somatic stem cells. Kim et al. matic hernia, Parkinson’s disease and cancer with
(2009a,b) have shown that it is possible to generate encouraging results. Finally, their usefulness for iPS
iPS cells by transducing adult mouse neural stem cells generation is very likely to expand their future clinical
(NSCs) and human foetal NSCs with Oct-4 only. Fur- use even further.
thermore, when human adipose MSCs were used, they
were reprogrammed more efficiently than fibroblasts
(Sun et al. 2009). REFERENCES
Few reports have also emerged on the use of cord and Abdallah, B. M. & Kassem, M. 2008 Human mesenchymal
extra ES cells for iPS generation. Galende et al. used stem cells: from basic biology to clinical applications.
terminally differentiated amniotic fluid cells as Gene Ther. 15, 109–116. (doi:10.1038/sj.gt.3303067)

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

12 Review. Characteristics of stem cells H. Abdulrazzak et al.

Aboody, K. S. et al. 2000 Neural stem cells display extensive Broxmeyer, H. 2010 Umbilical cord transplantation: epilogue.
tropism for pathology in adult brain: evidence from Semin. Hematol. 47, 97 –103. (doi:10.1053/j.seminhema-
intracranial gliomas. Proc. Natl Acad. Sci. USA 97, tol.2009.10.002)
12 846 –12 851. (doi:10.1073/pnas.97.23.12846) Broxmeyer, H. E., Douglas, G. W., Hangoc, G., Cooper, S.,
Alaminos, M., Perez-Kohler, B., Garzon, I., Garcia-Honduvilla, Bard, J., English, D., Arny, M., Thomas, L. & Boyse,
N., Romero, B., Campos, A. & Bujan, J. 2010 Transdiffer- E. A. 1989 Human umbilical cord blood as a potential
entiation potentiality of human Wharton’s jelly stem cells source of transplantable hematopoietic stem/progenitor
towards vascular endothelial cells. J. Cell. Physiol. 223, cells. Proc. Natl Acad. Sci. USA 86, 3828–3832. (doi:10.
640–647. 1073/pnas.86.10.3828)
Almeida-Porada, G., El Shabrawy, D., Porada, C. & Zanjani, Brunstein, C. G., Setubal, D. C. & Wagner, J. E. 2007
E. 2002 Differentiative potential of human metanephric Expanding the role of umbilical cord blood transplan-
mesenchymal cells. Exp. Hematol. 30, 1454–1462. tation. Br. J. Haematol. 137, 20– 35.
(doi:10.1016/S0301-472X(02)00967-0) Cai, J. et al. 2010 Generation of human induced pluripotent
Alviano, F. et al. 2007 Term amniotic membrane is a high stem cells from umbilical cord matrix and amniotic
throughput source for multipotent mesenchymal stem membrane mesenchymal cells. J. Biol. Chem. 285,
cells with the ability to differentiate into endothelial cells 11 227–11 234. (doi:10.1074/jbc.M109.086389)
in vitro. BMC Dev. Biol. 7, 11– 11. (doi:10.1186/1471- Campagnoli, C., Roberts, I., Kumar, S., Bennett, P.,
213X-7-11) Bellantuono, I. & Fisk, N. 2001 Identification of mesench-
Andrews, P. W., Matin, M. M., Bahrami, A. R., Damjanov, I., ymal stem/progenitor cells in human first-trimester fetal
Gokhale, P. & Draper, J. S. 2005 Embryonic stem (ES) blood, liver, and bone marrow. Blood 98, 2396– 2402.
cells and embryonal carcinoma (EC) cells: opposite sides (doi:10.1182/blood.V98.8.2396)
of the same coin. Biochem. Soc. Trans. 33, 1526–1530. Cananzi, M., Atala, A. & De Coppi, P. 2009 Stem cells
Anzalone, R., Iacono, M. L., Corrao, S., Magno, F., Loria, T., derived from amniotic fluid: new potentials in regenerative
Cappello, F., Zummo, G., Farina, F. & La Rocca, G. 2010 medicine. Reprod. Biomed. Online 18(Suppl. 1), 17– 27.
New emerging potentials for human Wharton’s jelly Cargnoni, A. et al. 2009 Transplantation of allogeneic and
mesenchymal stem cells: immunological features and hep- xenogeneic placenta-derived cells reduces bleomycin-
atocyte-like differentiative capacity. Stem Cells Dev. 19, induced lung fibrosis. Cell Transplant. 18, 405–422.
423 –438. (doi:10.1089/scd.2009.0299) (doi:10.3727/096368909788809857)
Atala, A. 2009 Engineering organs. Curr. Opin. Biotechnol. Carraro, G. et al. 2008 Human amniotic fluid stem cells can
20, 575 –592. (doi:10.1016/j.copbio.2009.10.003) integrate and differentiate into epithelial lung lineages.
Bailo, M. et al. 2004 Engraftment potential of human amnion Stem Cells 26, 2902–2911. (doi:10.1634/stemcells.2008-
and chorion cells derived from term placenta. Transplan- 0090)
tation 78, 1439–1448. (doi:10.1097/01.TP.0000144606. Castrechini, N. M., Murthi, P., Gude, N. M., Erwich, J. J.,
84234.49) Gronthos, S., Zannettino, A., Brennecke, S. P. & Kalionis,
Ballen, K. K. 2005 New trends in umbilical cord blood B. 2010 Mesenchymal stem cells in human placental chor-
transplantation. Blood 105, 3786–3792. (doi:10.1182/ ionic villi reside in a vascular niche. Placenta 31, 203–212.
blood-2004-10-4125) (doi:10.1016/j.placenta.2009.12.006)
Bankiewicz, K. S., Palmatier, M., Plunkett, R. J., Cummins, Chambers, I. et al. 2007 Nanog safeguards pluripotency and
A. & Oldfield, E. H. 1994 Reversal of hemiparkinsonian mediates germline development. Nature 450, 1230– 1234.
syndrome in nonhuman primates by amnion implantation (doi:10.1038/nature06403)
into caudate nucleus. J. Neurosurg. 81, 869 –876. Chan, J., O’Donoghue, K., de la Fuente, J., Roberts, I. A.,
Battula, V. L. et al. 2007 Human placenta and bone marrow Kumar, S., Morgan, J. E. & Fisk, N. M. 2005 Human
derived MSC cultured in serum-free, b-FGF-containing fetal mesenchymal stem cells as vehicles for gene delivery.
medium express cell surface frizzled-9 and SSEA-4 and Stem Cells 23, 93– 102. (doi:10.1634/stemcells.2004-0138)
give rise to multilineage differentiation. Differentiation Chan, J. et al. 2006 Galectin-1 induces skeletal muscle differ-
75, 279 –291. (doi:10.1111/j.1432-0436.2006.00139.x) entiation in human fetal mesenchymal stem cells and
Bhatia, M., Wang, J., Kapp, U., Bonnet, D. & Dick, J. 1997 increases muscle regeneration. Stem Cells 24, 1879– 1891.
Purification of primitive human hematopoietic cells (doi:10.1634/stemcells.2005-0564)
capable of repopulating immune-deficient mice. Proc. Chan, J., Waddington, S., O’Donoghue, K., Kurata, H.,
Natl Acad. Sci. USA 94, 5320–5325. (doi:10.1073/pnas. Guillot, P., Gotherstrom, C., Themis, M., Morgan, J. &
94.10.5320) Fisk, N. 2007 Widespread distribution and muscle differen-
Bieback, K., Kern, S., Kluter, H. & Eichler, H. 2004 Critical tiation of human fetal mesenchymal stem cells after
parameters for the isolation of mesenchymal stem cells intrauterine transplantation in dystrophic mdx mouse.
from umbilical cord blood. Stem Cells 22, 625–634. Stem Cells 25, 875–884. (doi:10.1634/stemcells.2006-0694)
(doi:10.1634/stemcells.22-4-625) Chiavegato, A. et al. 2007 Human amniotic fluid-derived stem
Bolisetty, S., Koh, T. H. H. G., Hammond, S., Panaretto, K. & cells are rejected after transplantation in the myocardium
Whitehall, J. 2002 Correlation of umbilical cord weight with of normal, ischemic, immuno-suppressed or immuno-
birth weight. Arch. Dis. Child Fetal Neonatal Ed. 86, F140. deficient rat. J. Mol. Cell. Cardiol. 42, 746–759. (doi:10.
(doi:10.1136/fn.86.2.F140-a) 1016/j.yjmcc.2006.12.008)
Brodeur, A., Wirth, D., Franklin, C., Reneker, L., Miner, J. & Chien, C., Yen, B., Lee, F., Lai, T., Chen, Y., Chan, S. &
Phillips, C. 2007 Type I collagen glomerulopathy: post- Huang, H. 2006 In vitro differentiation of human
natal collagen deposition follows glomerular maturation. placenta-derived multipotent cells into hepatocyte-like
Kidney Int. 71, 985 –993. (doi:10.1038/sj.ki.5002173) cells. Stem Cells 24, 1759–1768. (doi:10.1634/stemcells.
Brooke, G. et al. 2009 Manufacturing of human placenta- 2005-0521)
derived mesenchymal stem cells for clinical trials. Cipriani, S., Bonini, D., Marchina, E., Balgkouranidou, I.,
Br. J. Haematol. 144, 571 –579. (doi:10.1111/j.1365- Caimi, L., Grassi Zucconi, G. & Barlati, S. 2007 Mesench-
2141.2008.07492.x) ymal cells from human amniotic fluid survive and migrate

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 13

after transplantation into adult rat brain. Cell Biol. Int. Fu, Y.-S., Cheng, Y.-C., Lin, M.-Y. A., Cheng, H., Chu, P.-M.,
31, 845 –850. (doi:10.1016/j.cellbi.2007.01.037) Chou, S.-C., Shih, Y.-H., Ko, M.-H. & Sung, S. 2006 Con-
Clapp, D. W., Freie, B., Lee, W. H. & Zhang, Y. Y. 1995 Mol- version of human umbilical cord mesenchymal stem cells in
ecular evidence that in situ-transduced fetal liver Wharton’s jelly to dopaminergic neurons in vitro: potential
hematopoietic stem/progenitor cells give rise to medullary therapeutic application for Parkinsonism. Stem Cells 24,
hematopoiesis in adult rats. Blood 86, 2113 –2122. 115– 124. (doi:10.1634/stemcells.2005-0053)
Crane, J. & Cheung, S. 1988 An embryogenic model to Fuchs, J. R., Kaviani, A., Oh, J.-T., LaVan, D., Udagawa, T.,
explain cytogenetic inconsistencies observed in chorionic Jennings, R. W., Wilson, J. M. & Fauza, D. O. 2004 Dia-
villus versus fetal tissue. Prenat. Diagn. 8, 119– 129. phragmatic reconstruction with autologous tendon
(doi:10.1002/pd.1970080206) engineered from mesenchymal amniocytes. J. Pediatr.
Cumano, A., Ferraz, J. C., Klaine, M., Di Santo, J. P. & Surg. 39, 834– 838. (doi:10.1016/j.jpedsurg.2004.02.014)
Godin, I. 2001 Intraembryonic, but not yolk sac hemato- Fujimoto, K. L., Miki, T., Liu, L. J., Hashizume, R., Strom,
poietic precursors, isolated before circulation, provide S. C., Wagner, W. R., Keller, B. B. & Tobita, K. 2009
long-term multilineage reconstitution. Immunity 15, Naive rat amnion-derived cell transplantation improved
477– 485. (doi:10.1016/S1074-7613(01)00190-X) left ventricular function and reduced myocardial scar of
Dan, Y., Riehle, K., Lazaro, C., Teoh, N., Haque, J., postinfarcted heart. Cell Transplant. 18, 477–486.
Campbell, J. & Fausto, N. 2006 Isolation of multipotent (doi:10.3727/096368909788809785)
progenitor cells from human fetal liver capable of differen- Fukuchi, Y., Nakajima, H., Sugiyama, D., Hirose, I.,
tiating into liver and mesenchymal lineages. Proc. Natl Kitamura, T. & Tsuji, K. 2004 Human placenta-derived
Acad. Sci. USA 103, 9912–9917. (doi:10.1073/pnas. cells have mesenchymal stem/progenitor cell potential.
0603824103) Stem Cells 22, 649–658. (doi:10.1634/stemcells.22-5-649)
Da Sacco, S., Sedrakyan, S., Boldrin, F., Giuliani, S., Galende, E. et al. 2009 Amniotic fluid cells are more efficiently
Parnigotto, P., Habibian, R., Warburton, D., De Filippo, reprogrammed to pluripotency than adult cells. Cell
R. E. & Perin, L. 2010 Human amniotic fluid as a potential Reprogram 12, 117–125. (doi:10.1089/cell.2009.0077)
new source of organ specific precursor cells for future regen- Gammaitoni, L. et al. 2004 Elevated telomerase activity and
erative medicine applications. J. Urol. 183, 1193–1200. minimal telomere loss in cord blood long-term cultures
(doi:10.1016/j.juro.2009.11.006) with extensive stem cell replication. Blood 103, 4440–
De Coppi, P. et al. 2007a Isolation of amniotic stem cell lines 4448. (doi:10.1182/blood-2003-09-3079)
with potential for therapy. Nat. Biotechnol. 25, 100–106. Giorgetti, A. et al. 2009 Generation of induced pluripotent
(doi:10.1038/nbt1274) stem cells from human cord blood using OCT4 and
De Coppi, P. et al. 2007b Amniotic fluid and bone marrow SOX2. Cell Stem Cell 5, 353–357. (doi:10.1016/j.stem.
derived mesenchymal stem cells can be converted to 2009.09.008)
smooth muscle cells in the cryo-injured rat bladder and Giorgetti, A., Montserrat, N., Rodriguez-Piza, I., Azqueta, C.,
prevent compensatory hypertrophy of surviving smooth Veiga, A. & Izpisua Belmonte, J. C. 2010 Generation of
muscle cells. J. Urol. 177, 369 –376. induced pluripotent stem cells from human cord blood
Degistirici, O., Jager, M. & Knipper, A. 2008 Applicability of cells with only two factors: Oct4 and Sox2. Nat. Protoc.
cord blood-derived unrestricted somatic stem cells in tissue 5, 811–820. (doi:10.1038/nprot.2010.16)
engineering concepts. Cell Prolif. 41, 421 –440. (doi:10. Gosden, C. & Brock, D. J. 1978 Combined use of alphafeto-
1111/j.1365-2184.2008.00536.x) protein and amniotic fluid cell morphology in early
de la Fuente, J., Fisk, N., O’Donoghue, K., Chan, J., Kumar, prenatal diagnosis of fetal abnormalities. J. Med. Genet.
S. & Roberts, I. 2003 a2b1 and a4b1 integrins mediate the 15, 262–270. (doi:10.1136/jmg.15.4.262)
homing of mesenchymal stem/progenitor cells during fetal Gotherstrom, C., Ringden, O., Tammik, C., Zetterberg, E.,
life. Haematol. J. 4, 13. Westgren, M. & Le Blanc, K. 2004 Immunologic properties
Delaney, C., Heimfeld, S., Brashem-Stein, C., Voorhies, H., of human fetal mesenchymal stem cells. Am. J. Obstet.
Manger, R. & Bernstein, I. 2010 Notch-mediated expan- Gynecol. 190, 239–245. (doi:10.1016/j.ajog.2003.07.022)
sion of human cord blood progenitor cells capable of Grounds, M. D., Radley, H. G., Lynch, G. S., Nagaraju, K. &
rapid myeloid reconstitution. Nat. Med. 16, 232–236. De Luca, A. 2008 Towards developing standard operating
(doi:10.1038/nm.2080) procedures for pre-clinical testing in the mdx mouse model
Delo, D. et al. 2010 Calcification after myocardial infarction is of Duchenne muscular dystrophy. Neurobiol. Dis. 31,
independent of amniotic fluid stem cell injection. 1–19. (doi:10.1016/j.nbd.2008.03.008)
Cardiovasc. Pathol. (doi:10.1089/scd.2008.0028) Gucciardo, L., Lories, R., Ochsenbein-Kolble, N., Done, E.,
Ditadi, A. et al. 2009 Human and murine amniotic fluid Zwijsen, A. & Deprest, J. 2009 Fetal mesenchymal stem
c-Kitþ Lin2 cells display hematopoietic activity. Blood cells: isolation, properties and potential use in perinatology
113, 3953–3960. (doi:10.1182/blood-2008-10-182105) and regenerative medicine. Br. J. Obstet. Gynaecol. 116,
Elwan, M. A. & Sakuragawa, N. 1997 Evidence for synthesis 166– 172.
and release of catecholamines by human amniotic epi- Guillot, P. V., O’Donoghue, K., Kurata, H. & Fisk, N. M.
thelial cells. Neuroreport 8, 3435 –3438. (doi:10.1097/ 2006 Fetal stem cells: betwixt and between.
00001756-199711100-00004) Semin. Reprod. Med. 24, 340– 347. (doi:10.1055/s-2006-
Ersek, A., Pixley, J., Goodrich, A., Porada, C., 952149)
Almeida-Porada, G., Thain, D. & Zanjani, E. 2010 Persist- Guillot, P. V., Cui, W., Fisk, N. M. & Polak, D. J. 2007a Stem
ent circulating human insulin in sheep transplanted in cell differentiation and expansion for clinical applications
utero with human mesenchymal stem cells. Exp. Hematol. of tissue engineering. J. Cell. Mol. Med. 11, 935–944.
38, 311 –320. (doi:10.1016/j.exphem.2010.02.005) (doi:10.1111/j.1582-4934.2007.00106.x)
Fong, C. Y., Richards, M., Manasi, N., Biswas, A. & Bongso, Guillot, P. V., Gotherstrom, C., Chan, J., Kurata, H. & Fisk,
A. 2007 Comparative growth behaviour and characteriz- N. M. 2007b Human first-trimester fetal MSC express
ation of stem cells from human Wharton’s jelly. Reprod. pluripotency markers and grow faster and have longer
Biomed. Online 15, 708 –718. (doi:10.1016/S1472- telomeres than adult MSC. Stem Cells 25, 646–654.
6483(10)60539-1) (doi:10.1634/stemcells.2006-0208)

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

14 Review. Characteristics of stem cells H. Abdulrazzak et al.

Guillot, P. et al. 2008a Intrauterine transplantation of human Ilancheran, S., Moodley, Y. & Manuelpillai, U. 2009 Human
fetal mesenchymal stem cells from first-trimester blood fetal membranes: a source of stem cells for tissue regener-
repairs bone and reduces fractures in osteogenesis ation and repair? Placenta 30, 2–10. (doi:10.1016/j.
imperfecta mice. Blood 111, 1717– 1725. (doi:10.1182/ placenta.2008.09.009)
blood-2007-08-105809) Insausti, C., Blanquer, M., Bleda, P., Iniesta, P., Majado, M.,
Guillot, P., Cook, H., Pusey, C., Fisk, N., Harten, S., Moss, J., Castellanos, G. & Moraleda, J. 2010 The amniotic
Shore, I. & Bou-Gharios, G. 2008b Transplantation of membrane as a source of stem cells. Histol. Histopathol. 25,
human fetal mesenchymal stem cells improves glomerulo- 91–98.
pathy in a collagen type I alpha 2-deficient mouse. in‘t Anker, P. S. et al. 2003a Mesenchymal stem cells in
J. Pathol. 214, 627– 636. (doi:10.1002/path.2325) human second-trimester bone marrow, liver, lung, and
Habich, A., Jurga, M., Markiewicz, I., Lukomska, B., spleen exhibit a similar immunophenotype but a hetero-
Bany-Laszewicz, U. & Domanska-Janik, K. 2006 Early geneous multilineage differentiation potential.
appearance of stem/progenitor cells with neural-like Haematologica 88, 845–852.
characteristics in human cord blood mononuclear fraction in‘t Anker, P. S., Scherjon, S. A., Kleijburg-van der Keur, C.,
cultured in vitro. Exp. Hematol. 34, 914 –925. (doi:10. Noort, W. A., Claas, F. H., Willemze, R., Fibbe, W. E. &
1016/j.exphem.2006.03.010) Kanhai, H. H. 2003b Amniotic fluid as a novel source of
Harrison, D., Zhong, R., Jordan, C., Lemischka, I. & Astle, C. mesenchymal stem cells for therapeutic transplantation.
1997 Relative to adult marrow, fetal liver repopulates Blood 102, 1548–1549.
nearly five times more effectively long-term than in‘t Anker, P. S., Scherjon, S. A., Kleijburg-van der Keur, C.,
short-term. Exp. Hematol. 25, 293– 297. de Groot-Swings, G. M., Claas, F. H., Fibbe, W. E. &
Horwitz, E. M., Gordon, P. L., Koo, W. K. K., Marx, J. C., Kanhai, H. H. 2004 Isolation of mesenchymal stem
Neel, M. D., McNall, R. Y., Muul, L. & Hofmann, T. cells of fetal or maternal origin from human placenta.
2002 Isolated allogeneic bone marrow-derived mesenchy- Stem Cells 22, 1338–1345. (doi:10.1634/stemcells.2004-
mal cells engraft and stimulate growth in children with 0058)
osteogenesis imperfecta: implications for cell therapy of Jaenisch, R. 2009 Stem cells, pluripotency and nuclear
bone. Proc. Natl Acad. Sci. USA 99, 8932–8937. (doi:10. reprogramming. J. Thromb. Haemost. 7(Suppl. 1), 21–23.
1073/pnas.132252399) Javazon, E., Beggs, K. & Flake, A. 2004 Mesenchymal stem
Hsieh, J. Y., Fu, Y. S., Chang, S. J., Tsuang, Y. H. & Wang, cells: paradoxes of passaging. Exp. Hematol. 32,
H. W. 2010 Functional module analysis reveals differential 414–425. (doi:10.1016/j.exphem.2004.02.004)
osteogenic and stemness potentials in human mesenchymal Jomura, S., Uy, M., Mitchell, K., Dallasen, R., Bode, C. & Xu,
stem cells from bone marrow and Wharton’s jelly of umbi- Y. 2007 Potential treatment of cerebral global ischemia
lical cord. Stem Cells Dev. (doi:10.1089/scd.2009.0485) with Oct-4þ umbilical cord matrix cells. Stem Cells 25,
Hu, Y., Liao, L., Wang, Q., Ma, L., Ma, G., Jiang, X. & Zhao, 98 –106. (doi:10.1634/stemcells.2006-0055)
R. C. 2003 Isolation and identification of mesenchymal Jones, H. M., Nathrath, M., Thomas, R. M., Edelman, P.,
stem cells from human fetal pancreas. J. Lab. Clin. Med. Rodeck, C. H. & Linch, D. C. 1994 The effects of gestation
141, 342 –349. (doi:10.1016/S0022-2143(03)00022-2) on circulating progenitor cells. Br. J. Haematol. 87,
Huang, H. 2007 Isolation of human placenta-derived multi- 637–639. (doi:10.1111/j.1365-2141.1994.tb08329.x)
potent cells and in vitro differentiation into hepatocyte- Kakishita, K., Elwan, M. A., Nakao, N., Itakura, T. &
like cells. Curr. Protoc. Stem Cell Biol. 1, 1E.1. (doi:10. Sakuragawa, N. 2000 Human amniotic epithelial cells pro-
1002/9780470151808.sc01e01s1) duce dopamine and survive after implantation into the
Huang, G. T., Gronthos, S. & Shi, S. 2009 Mesenchymal stem striatum of a rat model of Parkinson’s disease: a potential
cells derived from dental tissues vs. those from other source of donor for transplantation therapy. Exp. Neurol.
sources: their biology and role in regenerative medicine. 165, 27–34. (doi:10.1006/exnr.2000.7449)
J. Dent. Res. 88, 792 –806. Kakishita, K., Nakao, N., Sakuragawa, N. & Itakura, T. 2003
Huangfu, D., Osafune, K., Maehr, R., Guo, W., Eijkelenboom, Implantation of human amniotic epithelial cells prevents
A., Chen, S., Muhlestein, W. & Melton, D. 2008 Induction the degeneration of nigral dopamine neurons in rats with
of pluripotent stem cells from primary human fibroblasts 6-hydroxydopamine lesions. Brain Res. 980, 48–56.
with only Oct4 and Sox2. Nat. Biotechnol. 26, (doi:10.1016/S0006-8993(03)02875-0)
1269– 1275. (doi:10.1038/nbt.1502) Karahuseyinoglu, S., Cinar, O., Kilic, E., Kara, F., Akay,
Huss, R. 2000 Isolation of primary and immortalized CD34- G. G., Demiralp, D. O., Tukun, A., Uckan, D. & Can, A.
hematopoietic and mesenchymal stem cells from various 2007 Biology of stem cells in human umbilical cord
sources. Stem Cells 18, 1 –9. (doi:10.1634/stemcells.18-1-1) stroma: in situ and in vitro surveys. Stem Cells 25,
Hwang, W. Y. K., Samuel, M., Tan, D., Koh, L. P., Lim, W. & 319–331. (doi:10.1634/stemcells.2006-0286)
Linn, Y. C. 2007 A meta-analysis of unrelated donor umbi- Kaviani, A., Perry, T. E., Dzakovic, A., Jennings, R. W.,
lical cord blood transplantation versus unrelated donor Ziegler, M. M. & Fauza, D. O. 2001 The amniotic fluid
bone marrow transplantation in adult and pediatric as a source of cells for fetal tissue engineering. J. Pediatr.
patients. Biol. Blood Marrow Transplant. 13, 444–453. Surg. 36, 1662– 1665. (doi:10.1053/jpsu.2001.27945)
(doi:10.1016/j.bbmt.2006.11.005) Kennea, N. et al. 2009 Differentiation of human fetal mesench-
Igura, K., Zhang, X., Takahashi, K., Mitsuru, A., Yamaguchi, ymal stem cells into cells with an oligodendrocyte
S. & Takashi, T. A. 2004 Isolation and characterization of phenotype. Cell Cycle 8, 1069–1079.
mesenchymal progenitor cells from chorionic villi of human Khan, A. A. et al. 2010 Human fetal liver derived stem cell
placenta. Cytotherapy 6, 543 –553. (doi:10.1080/ transplantation as supportive modality in the management
14653240410005366-1) of end stage decompensated liver cirrhosis. Cell Transplant.
Ilancheran, S., Michalska, A., Peh, G., Wallace, E. M., Pera, 19, 409–418.
M. & Manuelpillai, U. 2007 Stem cells derived from Kim, J., Lee, Y., Kim, H., Hwang, K. J., Kwon, H. C., Kim,
human fetal membranes display multilineage differen- S. K., Cho, D. J., Kang, S. G. & You, J. 2007 Human
tiation potential. Biol. Reprod. 77, 577– 588. (doi:10. amniotic fluid-derived stem cells have characteristics of
1095/biolreprod.106.055244) multipotent stem cells. Cell Prolif. 40, 75 –90.

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 15

Kim, S. M. et al. 2008 Gene therapy using TRAIL-secreting Lim, F. T. H., Kanhai, H. H. H. & Falkenburg, J. H. F. 2005
human umbilical cord blood-derived mesenchymal stem Characterization of the human CD34þ hematopoietic pro-
cells against intracranial glioma. Cancer Res. 68, genitor cell compartment during the second trimester of
9614–9623. (doi:10.1158/0008-5472.CAN-08-0451) pregnancy. Haematologica 90, 173–179.
Kim, J. et al. 2009a Oct4-induced pluripotency in adult Liu, T., Wu, J., Huang, Q., Hou, Y., Jiang, Z., Zang, S. &
neural stem cells. Cell 136, 411 –419. (doi:10.1016/j.cell. Guo, L. 2008 Human amniotic epithelial cells ameliorate
2009.01.023) behavioral dysfunction and reduce infarct size in the
Kim, J. B., Greber, B., Arauzo-Bravo, M. J., Meyer, J., Park, rat middle cerebral artery occlusion model. Shock 29,
K. I., Zaehres, H. & Scholer, H. R. 2009b Direct reprogram- 603– 611.
ming of human neural stem cells by OCT4. Nature 461, Lo, B. & Parham, L. 2009 Ethical issues in stem cell research.
643– 649. (doi:10.1038/nature08436) Endocr. Rev. 30, 204–213. (doi:10.1210/er.2008-0031)
Klimanskaya, I., Chung, Y., Becker, S., Lu, S. J. & Lanza, R. Loebinger, M. & Janes, S. 2010 Stem cells as vectors for anti-
2007 Derivation of human embryonic stem cells from single tumour therapy. Thorax 65, 362–369. (doi:10.1136/thx.
blastomeres. Nat. Protoc. 2, 1963– 1972. (doi:10.1038/ 2009.128025)
nprot.2007.274) Lund, R. D. et al. 2007 Cells isolated from umbilical cord
Koizumi, N. J., Inatomi, T. J., Sotozono, C. J., Fullwood, tissue rescue photoreceptors and visual functions in a
N. J., Quantock, A. J. & Kinoshita, S. 2000 Growth rodent model of retinal disease. Stem Cells 25, 602–611.
factor mRNA and protein in preserved human amniotic (doi:10.1634/stemcells.2006-0308)
membrane. Curr. Eye. Res. 20, 173 –177. MacKenzie, T. C., Campagnoli, C., Almeida-Porada, G., Fisk,
Kolambkar, Y. M., Peister, A., Soker, S., Atala, A. & N. M. & Flake, A. W. 2001 Circulating human fetal stro-
Guldberg, R. E. 2007 Chondrogenic differentiation of mal cells engraft and differentiate in multiple tissues
amniotic fluid-derived stem cells. J. Mol. Histol. 38, following transplantation into pre-immune fetal lambs.
405– 413. (doi:10.1007/s10735-007-9118-1) Blood 98, 798.
Kong, X.-Y. et al. 2008 Transplantation of human amniotic Maherali, N. et al. 2007 Directly reprogrammed fibroblasts
cells exerts neuroprotection in MPTP-induced Parkinson show global epigenetic remodeling and widespread tissue
disease mice. Brain Res. 1205, 108 –115. (doi:10.1016/j. contribution. Cell Stem Cell 1, 55 –70. (doi:10.1016/j.
brainres.2008.02.040) stem.2007.05.014)
Kubo, M., Sonoda, Y., Muramatsu, R. & Usui, M. 2001 Manzur, A. Y. & Muntoni, F. 2009 Diagnosis and new treat-
Immunogenicity of human amniotic membrane in exper- ments in muscular dystrophies. J. Neurol. Neurosurg.
imental xenotransplantation. Invest. Ophthalmol. Vis. Psychiatry 80, 706–714.
Sci. 42, 1539– 1546. Marcus, A. & Woodbury, D. 2008 Fetal stem cells from extra-
Kucia, M., Zuba-Surma, E., Wysoczynski, M., Dobrowolska, H., embryonic tissues: do not discard. J. Cell. Mol. Med. 12,
Reca, R., Ratajczak, J. & Ratajczak, M. 2006 Physiological 730– 742. (doi:10.1111/j.1582-4934.2008.00221.x)
and pathological consequences of identification of very Mazurier, F., Doedens, M., Gan, O. & Dick, J. 2003
small embryonic like (VSEL) stem cells in adult bone Rapid myeloerythroid repopulation after intrafemoral
marrow. J. Physiol. Pharmacol. 57(Suppl. 5), 5–18. transplantation of NOD– SCID mice reveals a new class
Kucia, M. et al. 2007 Morphological and molecular character- of human stem cells. Nat. Med. 9, 959–963. (doi:10.
ization of novel population of CXCR4þ SSEA-4þ Oct-4þ 1038/nm886)
very small embryonic-like cells purified from human cord McElreavey, K., Irvine, A., Ennis, K. & McLean, W. 1991 Iso-
blood: preliminary report. Leukemia 21, 297–303. lation, culture and characterisation of fibroblast-like cells
(doi:10.1038/sj.leu.2404470) derived from the Wharton’s jelly portion of human umbi-
Kunisaki, S. M., Jennings, R. W. & Fauza, D. O. 2006 Fetal lical cord. Biochem. Soc. Trans. 19, 29S.
cartilage engineering from amniotic mesenchymal progeni- McGrath, K. & Palis, J. 2008 Ontogeny of erythropoiesis in
tor cells. Stem Cells Dev. 15, 245 –253. (doi:10.1089/scd. the mammalian embryo. Curr. Top. Dev. Biol. 82, 1 –22.
2006.15.245) (doi:10.1016/S0070-2153(07)00001-4)
Kunisaki, S. M., Armant, M., Kao, G. S., Stevenson, K., Kim, Miao, Z., Jin, J., Chen, L., Zhu, J., Huang, W., Zhao, J., Qian,
H. & Fauza, D. O. 2007 Tissue engineering from human H. & Zhang, X. 2006 Isolation of mesenchymal stem cells
mesenchymal amniocytes: a prelude to clinical trials. from human placenta: comparison with human bone
J. Pediatr. Surg. 42, 974 –979. (doi:10.1016/j.jpedsurg. marrow mesenchymal stem cells. Cell Biol. Int. 30, 681–
2007.01.031) 687. (doi:10.1016/j.cellbi.2006.03.009)
Kyo, S. et al. 1997 Expression of telomerase activity in human Miki, T. & Strom, S. 2006 Amnion-derived pluripotent/multi-
chorion. Biochem. Biophys. Res. Commun. 241, 498– 503. potent stem cells. Stem Cell Rev. 2, 133–142. (doi:10.1007/
(doi:10.1006/bbrc.1997.7767) s12015-006-0020-0)
Le Blanc, K. et al. 2005 Fetal mesenchymal stem-cell engraft- Miki, T., Lehmann, T., Cai, H., Stolz, D. & Strom, S. 2005
ment in bone after in utero transplantation in a patient Stem cell characteristics of amniotic epithelial cells. Stem
with severe osteogenesis imperfecta. Transplantation 79, Cells 23, 1549–1559. (doi:10.1634/stemcells.2004-0357)
1607–1614. (doi:10.1097/01.TP.0000159029.48678.93) Miki, T., Mitamura, K., Ross, M., Stolz, D. & Strom, S. 2007
Le Blanc, K. et al. 2008 Mesenchymal stem cells for treatment Identification of stem cell marker-positive cells by immuno-
of steroid-resistant, severe, acute graft-versus-host disease: fluorescence in term human amnion. J. Reprod. Immunol.
a phase II study. Lancet 371, 1579–1586. (doi:10.1016/ 75, 91 –96. (doi:10.1016/j.jri.2007.03.017)
S0140-6736(08)60690-X) Miki, T., Marongiu, F., Dorko, K., Ellis, E. & Strom, S. 2010
Li, C., Zhang, W., Jiang, X. & Mao, N. 2007 Human-placenta- Isolation of amniotic epithelial stem cells. Curr. Protoc.
derived mesenchymal stem cells inhibit proliferation and Stem Cell Biol. 12, 1E.3.1–1E.3.10. (doi:10.1002/
function of allogeneic immune cells. Cell Tissue Res. 9780470151808.sc01e03s12)
330, 437 –446. (doi:10.1007/s00441-007-0504-5) Mitchell, K. E. et al. 2003 Matrix cells from Wharton’s jelly
Li, C. et al. 2009 Pluripotency can be rapidly and efficiently form neurons and glia. Stem Cells 21, 50– 60.
induced in human amniotic fluid-derived cells. Hum. Mol. Moon, J. H., Lee, J. R., Jee, B. C., Suh, C. S., Kim, S. H., Lim,
Genet. 18, 4340– 4349. (doi:10.1093/hmg/ddp386) H. J. & Kim, H. K. 2008 Successful vitrification of human

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

16 Review. Characteristics of stem cells H. Abdulrazzak et al.

amnion-derived mesenchymal stem cells. Hum. Reprod. villi of human placenta. Cytotherapy 10, 690–697. (doi:10.
23, 1760–1770. (doi:10.1093/humrep/den202) 1080/14653240802419310)
Mountford, J. 2008 Human embryonic stem cells: origins, Porada, C. D., Zanjani, E. D. & Almeida-Porad, G. 2006
characteristics and potential for regenerative therapy. Adult mesenchymal stem cells: a pluripotent population
Transfus. Med. 18, 1–12. (doi:10.1111/j.1365-3148.2007. with multiple applications. Curr. Stem Cell Res. Ther. 1,
00807.x) 365–369.
Musina, R., Bekchanova, E., Belyavskii, A., Grinenko, T. & Portmann-Lanz, C. B., Schoeberlein, A., Huber, A., Sager, R.,
Sukhikh, G. 2007 Umbilical cord blood mesenchymal Malek, A., Holzgreve, W. & Surbek, D. V. 2006 Placental
stem cells. Bull. Exp. Biol. Med. 143, 127 –131. (doi:10. mesenchymal stem cells as potential autologous graft for
1007/s10517-007-0032-z) pre- and perinatal neuroregeneration. Am. J. Obstet.
Nadri, S. & Soleimani, M. 2007 Comparative analysis of Gynecol. 194, 664–673. (doi:10.1016/j.ajog.2006.01.101)
mesenchymal stromal cells from murine bone marrow and Portmann-Lanz, C. B., Schoeberlein, A., Portmann, R.,
amniotic fluid. Cytotherapy 9, 729– 737. (doi:10.1080/ Mohr, S., Rollini, P., Sager, R. & Surbek, D. 2010 Turning
14653240701656061) placenta into brain: placental mesenchymal stem cells
Nava, S., Westgren, M., Jaksch, M., Tibell, A., Broomé, U., differentiate into neurons and oligodendrocytes.
Ericzon, B. & Sumitran-Holgersson, S. 2005 Characterization Am. J. Obstet. Gynecol. 202, 294.e1 –294.e11. (doi:10.
of cells in the developing human liver. Differentiation 73, 249– 1016/j.ajog.2009.10.893)
260. (doi:10.1111/j.1432-0436.2005.00019.x) Pranke, P., Hendrikx, J., Debnath, G., Alespeiti, G.,
O’Donoghue, K. & Fisk, N. M. 2004 Fetal stem cells. Best Rubinstein, P., Nardi, N. & Visser, J. 2005 Immunopheno-
Pract. Res. Clin. Obstet. Gynaecol. 18, 853 –875. type of hematopoietic stem cells from placental/umbilical
Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. 1996 cord blood after culture. Braz. J. Med. Biol. Res. 38,
Long-term lymphohematopoietic reconstitution by a 1775– 1789.
single CD34-low/negative hematopoietic stem cell. Science Prat-Vidal, C. et al. 2007 Umbilical cord blood-derived stem
273, 242 –245. (doi:10.1126/science.273.5272.242) cells spontaneously express cardiomyogenic traits. Trans-
Owen, M. & Friedenstein, A. J. 1988 Stromal stem cells: plant. Proc. 39, 2434–2437.
marrow-derived osteogenic precursors. Ciba Found. Prusa, A. R. & Hengstschlager, M. 2002 Amniotic fluid cells
Symp. 136, 42 –60. and human stem cell research: a new connection. Med.
Pan, H., Yang, D., Chiu, Y., Lai, S., Wang, Y., Chang, M. & Sci. Monit. 8, RA253–RA257.
Cheng, F. 2006 Enhanced regeneration in injured sciatic Prusa, A. R., Marton, E., Rosner, M., Bernaschek, G. &
nerve by human amniotic mesenchymal stem cell. J. Clin. Hengstschlager, M. 2003 Oct-4-expressing cells in
Neurosci. 13, 570–575. (doi:10.1016/j.jocn.2005.06.007) human amniotic fluid: a new source for stem cell
Pan, H., Cheng, F., Chen, C., Lai, S., Lee, C., Yang, D., research? Hum. Reprod. 18, 1489–1493. (doi:10.1093/
Chang, M. & Ho, S. 2007 Post-injury regeneration in rat humrep/deg279)
sciatic nerve facilitated by neurotrophic factors secreted Rachakatla, R. S., Marini, F., Weiss, M. L., Tamura, M. &
by amniotic fluid mesenchymal stem cells. J. Clin. Neuro- Troyer, D. 2007 Development of human umbilical cord
sci. 14, 1089–1098. (doi:10.1016/j.jocn.2006.08.008) matrix stem cell-based gene therapy for experimental
Pan, H. C., Yang, D. Y., Ho, S. P., Sheu, M. L., Chen, C. J., lung tumors. Cancer Gene Ther. 14, 828– 835. (doi:10.
Hwang, S. M., Chang, M. H. & Cheng, F. C. 2009 1038/sj.cgt.7701077)
Escalated regeneration in sciatic nerve crush injury Rebel, V., Miller, C., Eaves, C. & Lansdorp, P. 1996 The repo-
by the combined therapy of human amniotic fluid pulation potential of fetal liver hematopoietic stem cells
mesenchymal stem cells and fermented soybean extracts, in mice exceeds that of their liver adult bone marrow
Natto. J. Biomed. Sci. 16, 75. (doi:10.1186/1423-0127- counterparts. Blood 87, 3500–3507.
16-75) Rehni, A. K., Singh, N., Jaggi, A. S. & Singh, M. 2007 Amnio-
Pappa, K. & Anagnou, N. 2009 Novel sources of fetal stem tic fluid derived stem cells ameliorate focal cerebral
cells: where do they fit on the developmental continuum? ischaemia-reperfusion injury induced behavioural deficits
Regen. Med. 4, 423 –433. (doi:10.2217/rme.09.12) in mice. Behav. Brain Res. 183, 95– 100. (doi:10.1016/j.
Parolini, O. et al. 2008 Concise review: isolation and charac- bbr.2007.05.028)
terization of cells from human term placenta: outcome of Riekstina, U., Cakstina, I., Parfejevs, V., Hoogduijn, M.,
the first international Workshop on Placenta Derived Jankovskis, G., Muiznieks, I., Muceniece, R. & Ancans, J.
Stem Cells. Stem Cells 26, 300 –311. (doi:10.1634/stem- 2009 Embryonic stem cell marker expression pattern in
cells.2007-0594) human mesenchymal stem cells derived from bone
Parolini, O. et al. 2010 Toward cell therapy using placenta- marrow, adipose tissue, heart and dermis. Stem Cell Rev.
derived cells: disease mechanisms, cell biology, preclinical 5, 378–386. (doi:10.1007/s12015-009-9094-9)
studies, and regulatory aspects at the round table. Stem Rollini, P., Kaiser, S., Faes-van’t Hull, E., Kapp, U. &
Cells Dev. 19, 143 –154. (doi:10.1089/scd.2009.0404) Leyvraz, S. 2004 Long-term expansion of transplantable
Perin, L. et al. 2010 Protective effect of human amniotic fluid human fetal liver hematopoietic stem cells. Blood 103,
stem cells in an immunodeficient mouse model of acute 1166– 1170. (doi:10.1182/blood-2003-06-1815)
tubular necrosis. PLoS ONE 5, e9357. (doi:10.1371/jour- Roubelakis, M. G., Pappa, K. I., Bitsika, V., Zagoura, D.,
nal.pone.0009357) Vlahou, A., Papadaki, H. A., Antsaklis, A. & Anagnou,
Phillips, C., Pfeiffer, B., Luger, A. & Franklin, C. 2002 Novel N. P. 2007 Molecular and proteomic characterization of
collagen glomerulopathy in a homotrimeric type I collagen human mesenchymal stem cells derived from amniotic
mouse (oim). Kidney Int. 62, 383 –391. (doi:10.1046/j. fluid: comparison to bone marrow mesenchymal stem
1523-1755.2002.00451.x) cells. Stem Cells Dev. 16, 931–952. (doi:10.1089/scd.
Pittenger, M. F. et al. 1999 Multilineage potential of adult 2007.0036)
human mesenchymal stem cells. Science 284, 143–147. Sakuragawa, N. et al. 1997 Evidence for active acetylcholine
(doi:10.1126/science.284.5411.143) metabolism in human amniotic epithelial cells: applicable
Poloni, A. et al. 2008 Characterization and expansion of to intracerebral allografting for neurologic disease. Neurosci.
mesenchymal progenitor cells from first-trimester chorionic Lett. 232, 53–56. (doi:10.1016/S0304-3940(97)00570-3)

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

Review. Characteristics of stem cells H. Abdulrazzak et al. 17

Sakuragawa, N., Enosawa, S., Ishii, T., Thangavel, R., from human amniotic membranes and initiation of germ
Tashiro, T., Okuyama, T. & Suzuki, S. 2000 Human layers in vitro. Hum. Cell. 17, 125–130.
amniotic epithelial cells are promising transgene carriers Tamagawa, T., Oi, S., Ishiwata, I., Ishikawa, H. & Nakamura,
for allogeneic cell transplantation into liver. J. Hum. Y. 2007 Differentiation of mesenchymal cells derived from
Genet. 45, 171 –176. (doi:10.1007/s100380050205) human amniotic membranes into hepatocyte-like cells
Sakuragawa, N., Elwan, M. A., Uchida, S., Fujii, T. & in vitro. Hum. Cell. 20, 77–84. (doi:10.1111/j.1749-0774.
Kawashima, K. 2001 Non-neuronal neurotransmitters 2007.00032.x)
and neurotrophic factors in amniotic epithelial cells: Taylor, P. A., McElmurry, R. T., Lees, C. J., Harrison, D. E. &
expression and function in humans and monkey. Jpn Blazar, B. R. 2002 Allogenic fetal liver cells have a distinct
J. Pharmacol. 85, 20– 23. (doi:10.1254/jjp.85.20) competitive engraftment advantage over adult bone marrow
Schmidt, D. et al. 2006 Living autologous heart valves engin- cells when infused into fetal as compared with adult severe
eered from human prenatally harvested progenitors. combined immunodeficient recipients. Blood 99, 1870–
Circulation 114, I125– I131. (doi:10.1161/CIRCULATIO- 1872. (doi:10.1182/blood.V99.5.1870)
NAHA.105.001040) Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S., Waknitz,
Schoemans, H., Theunissen, K., Maertens, J., Boogaerts, M., M. A., Swiergiel, J. J., Marshall, V. S. & Jones, J. M.
Verfaillie, C. & Wagner, J. 2006 Adult umbilical cord 1998 Embryonic stem cell lines derived from human blasto-
blood transplantation: a comprehensive review. Bone cysts. Science 282, 1145– 1147. (doi:10.1126/science.282.
Marrow Transplant. 38, 83–93. (doi:10.1038/sj.bmt. 5391.1145)
1705403) Torricelli, F., Brizzi, L., Bernabei, P. A., Gheri, G., Di Lollo,
Secco, M. et al. 2008 Multipotent stem cells from umbilical S., Nutini, L., Lisi, E., Di Tommaso, M. & Cariati, E. 1993
cord: cord is richer than blood! Stem Cells 26, 146–150. Identification of hematopoietic progenitor cells in human
(doi:10.1634/stemcells.2007-0381) amniotic fluid before the 12th week of gestation.
Sessarego, N. et al. 2008 Multipotent mesenchymal stromal Ital. J. Anat. Embryol. 98, 119–126.
cells from amniotic fluid: solid perspectives for clinical Troyer, D. L. & Weiss, M. L. 2008 Wharton’s jelly-derived
application. Haematologica 93, 339 –346. (doi:10.3324/ cells are a primitive stromal cell population. Stem Cells
haematol.11869) 26, 591–599. (doi:10.1634/stemcells.2007-0439)
Siegel, N., Rosner, M., Hanneder, M., Valli, A. & Hengstschlager, Tsai, M. S., Lee, J. L., Chang, Y. J. & Hwang, S. M. 2004 Iso-
M. 2007 Stem cells in amniotic fluid as new tools to study lation of human multipotent mesenchymal stem cells from
human genetic diseases. Stem Cell Rev. 3, 256–264. (doi:10. second-trimester amniotic fluid using a novel two-stage
1007/s12015-007-9003-z) culture protocol. Hum. Reprod. 19, 1450– 1456. (doi:10.
Soncini, M., Vertua, E., Gibelli, L., Zorzi, F., Denegri, M., 1093/humrep/deh279)
Albertini, A., Wengler, G. S. & Parolini, O. 2007 Tsai, M.-S., Hwang, S.-M., Tsai, Y.-L., Cheng, F.-C., Lee,
Isolation and characterization of mesenchymal cells from J.-L. & Chang, J. 2006 Clonal amniotic fluid-derived
human fetal membranes. J. Tissue Eng. Regen. Med. 1, stem cells express characteristics of both mesenchymal
296– 305. (doi:10.1002/term.40) and neural stem cells. Biol. Reprod. 74, 545–551.
Spitalieri, P. et al. 2009 Identification of multipotent cyto- (doi:10.1095/biolreprod.105.046029)
trophoblast cells from human first trimester chorionic Tsai, M. et al. 2007 Functional network analysis of the tran-
villi. Cloning Stem Cells 11, 535 –556. (doi:10.1089/clo. scriptomes of mesenchymal stem cells derived from
2009.0046) amniotic fluid, amniotic membrane, cord blood, and
Streubel, B., Martucci-Ivessa, G., Fleck, T. & Bittner, R. E. bone marrow. Stem Cells 25, 2511–2523. (doi:10.1634/
1996 In vitro transformation of amniotic cells to muscle stemcells.2007-0023)
cells– background and outlook. Wien. Med. Wochenschr. Uchida, S., Inanaga, Y., Kobayashi, M., Hurukawa, S., Araie,
146, 216 –217. M. & Sakuragawa, N. 2000 Neurotrophic function of con-
Studeny, M., Marini, F. C., Dembinski, J. L., Zompetta, C., ditioned medium from human amniotic epithelial cells.
Cabreira-Hansen, M., Bekele, B. N., Champlin, R. E. & J. Neurosci. Res. 62, 585–590. (doi:10.1002/1097-
Andreeff, M. 2004 Mesenchymal stem cells: potential pre- 4547(20001115)62:4,585::AID-JNR13.3.0.CO;2-U)
cursors for tumor stroma and targeted-delivery vehicles Valli, A. et al. 2010 Embryoid body formation of human
for anticancer agents. J. Natl Cancer Inst. 96, 1593–1603. amniotic fluid stem cells depends on mTOR. Oncogene
Sudo, K., Kanno, M., Miharada, K., Ogawa, S., Hiroyama, T., 29, 966–977. (doi:10.1038/onc.2009.405)
Saijo, K. & Nakamura, Y. 2007 Mesenchymal progenitors Vaziri, H. et al. 2010 Spontaneous reversal of the developmen-
able to differentiate into osteogenic, chondrogenic, tal aging of normal human cells following transcriptional
and/or adipogenic cells in vitro are present in most reprogramming. Regen. Med. 5, 345–363. (doi:10.2217/
primary fibroblast-like cell populations. Stem Cells 25, rme.10.21)
1610–1617. (doi:10.1634/stemcells.2006-0504) Ventura, C. et al. 2007 Hyaluronan mixed esters of butyric
Sun, N. et al. 2009 Feeder-free derivation of induced pluri- and retinoic acid drive cardiac and endothelial fate in
potent stem cells from adult human adipose stem cells. term placenta human mesenchymal stem cells and enhance
Proc. Natl Acad. Sci. USA 106, 15 720 –15 725. (doi:10. cardiac repair in infarcted rat hearts. J. Biol. Chem. 282,
1073/pnas.0908450106) 14 243–14 252. (doi:10.1074/jbc.M609350200)
Takahashi, K. & Yamanaka, S. 2006 Induction of pluripotent Wagers, A. J., Christensen, J. L. & Weissman, I. L. 2002 Cell
stem cells from mouse embryonic and adult fibroblast cul- fate determination from stem cells. Gene Ther. 9, 606– 612.
tures by defined factors. Cell 126, 663 –676. (doi:10.1016/j. (doi:10.1038/sj.gt.3301717)
cell.2006.07.024) Wang, H.-S., Hung, S.-C., Peng, S.-T., Huang, C.-C., Wei,
Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, H.-M., Guo, Y.-J., Fu, Y.-S., Lai, M.-C. & Chen, C. 2004
T., Tomoda, K. & Yamanaka, S. 2007 Induction of pluripo- Mesenchymal stem cells in the Wharton’s jelly of the
tent stem cells from adult human fibroblasts by defined human umbilical cord. Stem Cells 22, 1330–1337.
factors. Cell 131, 861–872. (doi:10.1016/j.cell.2007.11.019) (doi:10.1634/stemcells.2004-0013)
Tamagawa, T., Ishiwata, I. & Saito, S. 2004 Establishment Wang, J. et al. 2003 SCID-repopulating cell activity of human
and characterization of a pluripotent stem cell line derived cord blood-derived CD34- cells assured by intra-bone

J. R. Soc. Interface
Downloaded from rsif.royalsocietypublishing.org on August 28, 2010

18 Review. Characteristics of stem cells H. Abdulrazzak et al.

marrow injection. Blood 101, 2924–2931. (doi:10.1182/ Yen, B. L., Chien, C.-C., Chen, Y.-C., Chen, J.-T., Huang,
blood-2002-09-2782) J.-S., Lee, F.-K. & Huang, I. 2008 Placenta-derived multi-
Wang, J., Wang, L., Wu, Y., Xiang, Y., Xie, C., Jia, B., potent cells differentiate into neuronal and glial cells in
Harrington, J. & McNiece, I. 2004 Mesenchymal stem/ vitro. Tissue Eng. Part A 14, 9–17. (doi:10.1089/ten.a.
progenitor cells in human umbilical cord blood as support 2006.0352)
for ex vivo expansion of CD34(þ) hematopoietic stem cells Yu, L. C., Wall, D. A., Sandler, E., Chan, K. W., Grayson, G. &
and for chondrogenic differentiation. Haematologica 89, Kletzel, M. 2001 Unrelated cord blood transplant experience
837 –844. by the pediatric blood and marrow transplant consortium.
Weiss, M. L. & Troyer, D. L. 2006 Stem cells in the umbilical Pediatr. Hematol. Oncol. 18, 235–245. (doi:10.1080/
cord. Stem Cell Rev. 2, 155 –162. (doi:10.1007/s12015-006- 088800101750238531)
0022-y) Yu, J. et al. 2007 Induced pluripotent stem cell lines derived
Weiss, M. L. et al. 2006 Human umbilical cord matrix from human somatic cells. Science 318, 1917 –1920.
stem cells: preliminary characterization and effect of (doi:10.1126/science.1151526)
transplantation in a rodent model of Parkinson’s Zhang, Y., Li, C. D., Jiang, X. X., Li, H. L., Tang, P. H. &
disease. Stem Cells 24, 781 –792. (doi:10.1634/stemcells. Mao, N. 2004 Comparison of mesenchymal stem cells
2005-0330) from human placenta and bone marrow. Chin. Med. J.
Weissman, I. & Shizuru, J. 2008 The origins of the (Engl) 117, 882–887.
identification and isolation of hematopoietic stem Zhang, Z. Y., Teoh, S. H., Chong, M. S., Schantz, J. T., Fisk,
cells, and their capability to induce donor-specific trans- N. M., Choolani, M. A. & Chan, J. 2009 Superior osteo-
plantation tolerance and treat autoimmune diseases. genic capacity for bone tissue engineering of fetal
Blood 112, 3543–3553. (doi:10.1182/blood-2008-08- compared with perinatal and adult mesenchymal stem
078220) cells. Stem Cells 27, 126–137. (doi:10.1634/stemcells.
Weissman, I., Anderson, D. & Gage, F. 2001 Stem and 2008-0456)
progenitor cells: origins, phenotypes, lineage commitments, Zhao, P., Ise, H., Hongo, M., Ota, M., Konishi, I. & Nikaido,
and transdifferentiations. Annu. Rev. Cell. Dev. Biol. 17, T. 2005 Human amniotic mesenchymal cells have some
387 –403. (doi:10.1146/annurev.cellbio.17.1.387) characteristics of cardiomyocytes. Transplantation 79,
Yao, Z., Qin, M., Liu, J., Chen, X. & Zhou, D. 2004 In vitro 528–535. (doi:10.1097/01.TP.0000149503.92433.39)
cultivation of human fetal pancreatic ductal stem cells Zhao, Y., Wang, H. & Mazzone, T. 2006 Identification of stem
and their differentiation into insulin-producing cells. cells from human umbilical cord blood with embryonic and
World J. Gastroenterol. 10, 1452–1456. hematopoietic characteristics. Exp. Cell Res. 312, 2454–
Yeh, Y. C. et al. 2010 Cellular cardiomyoplasty with 2464. (doi:10.1016/j.yexcr.2006.04.008)
human amniotic fluid stem cells: in vitro in vivo studies. Zuba-Surma, E. K., Klich, I., Greco, N., Laughlin, M. J.,
Tissue Eng. Part A 16, 1925– 1936. (doi:10.1089/ten.tea. Ratajczak, J. & Ratajczak, M. Z. 2010 Optimization of
2009.0728) isolation and further characterization of umbilical-cord-
Yen, B. L. et al. 2005 Isolation of multipotent cells from blood-derived very small embryonic/ epiblast-like stem
human term placenta. Stem Cells 23, 3–9. (doi:10.1634/ cells (VSELs). Eur. J. Haematol. 84, 34 –46. (doi:10.
stemcells.2004-0098) 1111/j.1600-0609.2009.01352.x)

J. R. Soc. Interface

Vous aimerez peut-être aussi