Sunteți pe pagina 1din 8

Placenta 35 (2014) 77e84

Contents lists available at ScienceDirect

Placenta
journal homepage: www.elsevier.com/locate/placenta

Current opinion

Can we fix it? Evaluating the potential of placental stem cells for the
treatment of pregnancy disorders
J.L. James*, S. Srinivasan, M. Alexander, L.W. Chamley
Department of Obstetrics and Gynaecology, University of Auckland, New Zealand

a r t i c l e i n f o a b s t r a c t

Article history: In pregnancy disorders such as pre-eclampsia, intrauterine growth restriction (IUGR) and recurrent
Accepted 22 December 2013 miscarriage a poorly functioning placenta is thought to be a major component of the disease process.
However, despite their prevalence, we currently have no way to fix dysfunctional placentae or directly
Keywords: treat these disorders. Over the past two decades our understanding of the role that stem cells play in
Placenta organ development and regeneration has expanded rapidly, and over the past 5 years the therapeutic use
Stem cell
of stem cells to both regenerate damaged tissues, and act as potent modulators of diseased microenvi-
Regenerative medicine
ronments, has become a reality in many organs including the heart, kidney, liver, skin and eye. Over its
Pre-eclampsia
IUGR
short lifespan the placenta undergoes rapid and continuous growth and differentiation, meaning that
placental ‘organogenesis’ only truly ends at delivery, and thus stem cells are likely to play important roles
in placental function for the duration of pregnancy. Two populations of stem cells exist in the placenta
that contribute to this on-going growth and differentiation: trophoblast stem cells and mesenchymal
stem cells. This review will address our current understanding of how each of these stem cell populations
contributes to successful placental function, how epithelial and mesenchymal stem cell populations are
being translated to the clinic in other fields, and whether these advances can teach us anything about
how placental stem cells could be used to fix faulty placentae in the future.
Ó 2013 Elsevier Ltd. All rights reserved.

1. Introduction significant and on-going part of medical practice in many different


medical fields in the future, how do we ensure that obstetric
In diseases of pregnancy such as intrauterine growth restriction medicine is not left behind? This review aims to address our cur-
(IUGR) a poorly functioning placenta is thought to be a major rent understanding of how stem cells contribute to successful
component of the disease process. However, despite our absolute placental function, how similar mesenchymal and epithelial stem
reliance on the placenta, we do not understand the basic biological cell populations are being translated to the clinic in other fields, and
processes that underpin placental formation, nor why these may whether these advances can teach us anything about how placental
fail, and we cannot effectively treat faulty placentae. Imagine that stem cells could be used to fix faulty placentae in the future.
we could fix the failing placenta by redirecting cell lineage differ-
entiation or, by implanting stem cells to replace poorly functioning
2. How do stem cells contribute to normal placental
parts of the placenta. To do this requires harvesting the enormous
function?
potential of stem cells to improve placental function. At present,
stem cell therapies to treat placental disorders seem like a far-
Following conception the newly fertilised oocyte undergoes a
fetched idea, but in reality there are currently hundreds of clin-
series of divisions producing equipotent daughter cells until at the
ical trials investigating the use of stem cells as therapeutics regis-
32/64 cell stage the trophectoderm separates from the inner cell
tered on the US clinical trial database (clinicaltrials.gov), and it is
mass. At nidation, following contact with the decidua the outer
likely that advances in stem cell biology will lead to major changes
trophectoderm layer begins to form the first primitive trophoblast
in medical care in the future. As stem cell therapies become a
populations e the primitive cytotrophoblast and primitive syncy-
tiotrophoblast [1]. As the early cytotrophoblast populations pro-
liferate they form invaginations into which mesenchymal cells
* Corresponding author. Department of Obstetrics and Gynaecology, FMHS,
University of Auckland, Private Bag 92019, Auckland Mail Centre, Auckland, New
derived from the differentiating inner cell mass invade, forming the
Zealand. Tel.: þ64 93737599. first placental villi at around 12 days post-fertilisation [1]. There-
E-mail address: j.james@auckland.ac.nz (J.L. James). fore, our current understanding is that the placenta is originally

0143-4004/$ e see front matter Ó 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.placenta.2013.12.010
78 J.L. James et al. / Placenta 35 (2014) 77e84

formed from two stem cell populations; 1) trophoblast stem cells growth media also adopt an endothelial-like cobblestone
(TSC) derived from the trophectoderm that go on to form all the morphology and up-regulate early endothelial lineage cell surface
differentiated trophoblast lineages of the placenta, and 2) mesen- markers including VEGFR2, but to date conditions have not been
chymal stem cells (MSC) that form all non-trophoblast cells of the established to induce the up-regulation of late endothelial markers
villous core. As the placenta continues to develop it undergoes an such as CD31 or vWF by cells derived from placental MSC [10].
intricate programme of organogenesis well in advance of the As placental MSC reside in a peri-vascular niche in the placenta
development of individual fetal organs. Indeed, the placenta is the throughout gestation, it is likely that they also continue to support
first fetal organ in which de novo blood vessel development occurs, the expansion of the placental vascular network by contributing to
with the first blood vessels evident in tertiary villi at just 15 days the process of angiogenesis, which takes over from vasculogenesis
post-fertilisation, a time when the embryo exists only as the three as the dominant mechanism for vascular development at around 32
ectodermal, endodermal and mesodermal layers and contains no days post-fertilisation [4,16,17]. This switch coincides with the
blood vessels [1,2]. onset of circulation between the placenta and fetus via the um-
bilical cord [17], thus opening up the possibility that both MSC
2.1. Mesenchymal stem cells originating in the placenta, and MSC circulating in fetal blood can
begin to contribute to placental vascular development from that
Mesenchymal Stem Cells (MSC) are multipotent cells that are time point. There are two forms of angiogenesis that contribute to
precursors to tissues of mesodermal lineages. MSCs were first iso- the placental vascular network. Branching angiogenesis, which
lated from bone marrow in 1974 [3], and have now been identified involves the proliferation and migration of endothelial cells to
in almost all tissues, including muscle, adipose tissue, lung, elongate and form lateral extensions from existing endothelial
Wharton’s Jelly, umbilical cord blood and placenta [4,5]. In order for tubes, leads to the formation of a branching vascular tree and is the
cells to be characterised as MSCs they must be capable of adhering predominant mechanism of angiogenesis in the placenta between
to plastic and differentiation into the three major mesenchymal weeks 6e24 of gestation. Branching angiogenesis results in the
lineages; adipocytes, chondrocytes or osteocytes [6]. MSCs must formation of terminal capillaries, which are evident as highly coiled
also express characteristic cell surface markers including CD73, blood vessels with a dilated lumen located close to the overlying
CD90, CD105 and HLA-A2 and lack expression of makers of other trophoblast to ensure efficient exchange between the maternal and
cell lineages including CD34, CD45, CD14 or CD11b, CD79a or CD19 fetal circulations [17]. After 24 weeks of gestation non-branching
and HLA-DR [6]. angiogenesis predominates, which results in the elongation of
Placental MSCs have been postulated to play an important role vascular capillary loops as a result of endothelial cell proliferation,
in placental development and function by developing the branch- and ensures that the placenta is able to keep up with fetal growth
ing villus structure of the placenta and contributing to vasculo- demands during the third trimester of pregnancy [17].
genesis and angiogenesis. Vasculogenesis (the de novo synthesis of
blood vessels) begins in the placenta with a change in morphology 2.2. Trophoblast stem cells
of MSCs at around 15e20 days post-fertilisation, which coincides
with the appearance of haemangiogenic stem cells that aggregate Trophoblasts are epithelial cells that are unique to the placenta.
into cell cords [7,8]. These haemangiogenic stem cells then differ- There are at least three mature subtypes of trophoblast in the hu-
entiate into CD34 positive endothelial cell cords in which primitive man placenta; cytotrophoblasts, extravillous trophoblasts and the
lumens are evident from 23 to 26 days post-fertilisation [7,8]. These syncytiotrophoblast, each of which have separate functions that are
vessels are connected by microvascular tubes, and by day 30 post- required to ensure pregnancy success. All of these three mature
fertilisation (six and a half weeks of gestation) a connecting trophoblast populations are derived initially from the trophecto-
network of larger vascular structures with clear lumens are evident derm lineage of the pre-implantation blastocyst. In the mouse, TSCs
within the placenta [7]. In an adult, vasculogenesis occurs during that give rise to all murine trophoblast types have been isolated
tissue remodelling, wound healing and tumour growth and in- from the trophectoderm, and as a result of experiments employing
volves the differentiation of existing bone marrow derived endo- these TSCs we have a good understanding of murine trophoblast
thelial progenitor cells into endothelial cells [9]. However, no bone differentiation [18]. However, the murine placenta is anatomically
marrow derived endothelial progenitor cells exist in the placenta at very different to the human placenta and murine trophoblast lin-
the beginning of pregnancy, and therefore placental vasculogenesis eages bear little resemblance to human trophoblast lineages [18].
occurs under very different circumstances. Instead, it has been Obtaining human TSCs would be of great value to better understand
proposed that placental vasculogenesis is initiated by the differ- how mature trophoblast populations are formed in the human
entiation of placental MSCs [8] (Fig. 1). placenta and ultimately how human TSCs may be used to fix faulty
MSC plasticity is influenced by the tissue of origin, and corre- placentae.
spondingly placental MSCs demonstrate limited potential to Traditionally villous cytotrophoblasts were termed ‘stem like
differentiate into unrelated cell types such as cardiomyocytes and cells’ that were believed to be multi-potent and give rise to all the
skeletal muscle, and are less efficient at differentiating into osteo- mature trophoblast lineages. However, many groups including ours
cytes and adipocytes than their bone marrow derived counterparts have presented evidence to challenge the multi-potent nature of
[10]. However, several MSC populations, including those derived villous cytotrophoblasts, and have instead suggested that different
from bone marrow and amniotic membrane, have demonstrated an cytotrophoblast sub-populations primed to differentiate into syn-
inherent predisposition to differentiate into an endothelial cytiotrophoblast or extravillous trophoblast exist [19e21]. Indeed, a
phenotype [11,12], and the pro-angiogenic environment generated feature of other epithelial stem cell populations is their generation
by trophoblast secretion of angiogenic factors such as VEGF family of highly proliferative progeny, referred to as transiently amplifying
members, angiopoietins, FGF and PDGF, strongly suggests that cells, that are intermediate between stem and terminally differ-
MSCs within placental villi are likely to have a similar predisposi- entiated cells. The transiently amplifying cells divide actively for a
tion [13]. Bone marrow-derived MSC treated with VEGF can be period of time to expand the pool of progenitor cells able to
induced to differentiate into endothelial-like cells that express both differentiate into the terminal cell lineages of the tissue, thus
vWF and Flt1 [14,15]. In a similar manner, MSC isolated from first increasing the number of differentiated progeny a stem cell can
trimester placental villi and cultured in commercial endothelial produce, and enabling the stem cell itself to divide infrequently and
J.L. James et al. / Placenta 35 (2014) 77e84 79

avoid the accumulation of replication-associated damage [22]. In trophoblast and mesenchymal lineages [17]. Whilst our under-
the human placenta, it is likely that villous cytotrophoblasts standing of the end-stage pathophysiology of pre-eclampsia and
represent such a transiently amplifying population, and consensus IUGR has come a long way in the past decade, there is still a huge
is now growing that a ‘true’ TSC population must exist that drives amount we do not understand about the root cause of these dis-
cytotrophoblast expansion. Despite this theoretical consensus, in orders, and understanding how stem cells contribute to normal and
practice, attempts by researchers to isolate and study human TSCs abnormal placentation has significant promise to rectify this.
in vitro have encountered many difficulties (reviewed in Ref. [18]) However, perhaps because the application of stem cell biology to
and to date a true human trophoblast stem cell remains elusive. the placenta has largely been a recent phenomenon, at present we
have almost no understanding of how either trophoblast or
3. Is placental stem cell function perturbed in pregnancy mesenchymal stem cell dysfunction may contribute to defects in
disorders? placentation.
The lack of a validated human TSC is an obvious road block to
In diseases of pregnancy such as pre-eclampsia and IUGR a understanding whether TSC deficiencies contribute to placental
poorly functioning placenta is thought to be a major component of diseases. However, even for MSCs, which can be readily isolated
the disease process. This placental dysfunction incorporates inad- from placentae throughout gestation, only a handful of studies
equate differentiation and function of cells derived from both the directly examining differences in placental MSC phenotype/

Fig. 1. Schematic diagram illustrating how MSC may contribute to placental vasculogenesis and angiogenesis during pregnancy. As no bone marrow derived endothelial progenitor
cells (EPC), which contribute to vasculogenesis in adult tissue, exist at the time of placental vasculogenesis, it has been hypothesised that early in pregnancy placental EPC/
endothelial cells are derived from placental MSC. (VSMC: vascular smooth muscle cells).
80 J.L. James et al. / Placenta 35 (2014) 77e84

function in pregnancy disorders have been published. All of these than engraftment into healthy cardiac tissue [32]. However,
studies highlight the potential importance of MSC in pregnancy engraftment rates are still low (0.1e3%), suggesting that thera-
disorders. Placental MSCs isolated from pre-eclamptic placentae peutic benefits are mediated by an indirect primary mechanism
have a more pro-inflammatory cytokine profile in comparison to rather than structural integration and differentiation into cardiac
those from normal placentae, and conditioned media from pre- myocytes [31,33].
eclamptic MSCs increased sFlt1 and macrophage migration inhib- It is becoming increasingly apparent that rather than just
itory factor (MIF) secretion by villous explants, indicating that directly contributing to cell regeneration, the therapeutic effects of
either free or exosome bound factors released from MSCs may MSC may arise as a result of their ability to modulate the local
induce a pre-eclamptic placental phenotype [23]. Defects are also environment, resulting in both the suppression of inflammation
observed in MSCs isolated from the decidua of pre-eclamptic and the promotion of vascularisation [34,35]. Preferential migra-
placentae, which produce higher levels of soluble ICAM-1, a cyto- tion of MSC appears to be mediated by the release of chemotactic
kine associated with inflammation, and SDF-1, a cytokine that signals from injured tissues, and MSC express a wide range of
stimulates MSC migration [24]. MSCs derived from the decidua of chemokine receptors (CCR1, CCR2, CCR5, CXCR1, CXCR4 and CXCR5)
pre-eclamptic pregnancies also show increased expression of miR- that appear to be important in site-directed migration of MSC
16, which has anti-angiogenic consequences as miR-16 over- [36,37]. Once at the site of injury, MSC have unique immunomod-
expressing MSCs produce less VEGF-A and endothelial cells ulatory properties that may reduce immune-mediated damage to
exposed to microparticles from these cells have a reduced capacity chronically inflamed tissues by altering the activities of different
to form vascular networks in vitro [25]. cells of the innate and adaptive immune systems (Fig. 2). Specif-
In IUGR pregnancies, endothelial progenitor cells derived from ically, MSC secrete soluble factors such as prostaglandins (PGE2),
cord blood have reduced vasculogenic capacity in comparison to indoleamine 2,3-dioxygenase (IDO) and nitric oxide (NO) that
those isolated from cord blood from normal pregnancies [26]. inhibit proliferation of T lymphocytes and down regulate T and B
Furthermore, endothelial colony forming cells from low birth lymphocyte production of IFNg and TNFa, while up-regulating their
weight pre-term babies have a reduced ability to migrate, prolif- production of IL-10, resulting in an anti-inflammatory environment
erate or form angiogenic tubes in vitro, and have increased [38e40]. Additionally, MSC inhibit the proliferation of Natural Killer
expression of anti-angiogenic genes such as thrombospondin-1 cells and modulate the immune profile of macrophages, both of
[27]. Such endothelial progenitor cells are also thought to exist in which are important in regulating the placental and uterine envi-
the placenta prior to the establishment of the feto-placental cir- ronment [41,42].
culation [2,28], and thus functional differences in fetal derived MSC promote vascularisation by the secretion of both pro-
endothelial progenitor populations may also hint at differences in angiogenic factors such as VEGF and TGFb that stimulate endo-
placental MSC function, particularly if such differences are a result thelial growth [43], and anti-apoptotic factors such as IGF-1 and
of genetic or epigenetic mechanisms common to both fetus and SFRP1 which may prevent endothelial cell death [44,45]. In
placenta. Therefore, it is surprising that differences in MSC between response to hypoxia-reperfusion injury, MSC stimulate nearby cells
normal and IUGR pregnancies have never been directly compared, to increase their expression of VEGF, which in turn causes MSC to
and this is an area which should be the focus of future research. up-regulate VEGF receptors 1 and 2 [46]. Concomitant expression of
these VEGF family members stimulates angiogenic organisation of
4. Stem cells and regenerative medicine e the future of endothelial tubes in capillary networks. Indeed, incubation of MSC-
therapeutic interventions? conditioned medium with cardiomyocytes subjected to hypoxia
reperfusion injury demonstrates a cardioprotective effect by
Over the past two decades our understanding of the role that reducing the severity of cell damage [47].
stem cells play in organ development and regeneration has
expanded rapidly, and over the past 5 years the therapeutic use of 4.2. Epithelial stem cells
stem cells to both regenerate damaged tissues, and act as potent
modulators of diseased microenvironments, has become a reality in Epithelial stem cells are responsible for the continuous regen-
many organs including the heart, kidney, liver, skin and eye. Whilst eration and repair of squamous epithelial layers in the skin, lung,
many adult stem cell populations exist in the body, this review will cornea, colon and glandular tissue. However, in contrast to the
focus on those directly applicable to the placenta e MSC and accelerating pace of MSC use in therapeutic medicine, there are less
epithelial stem cells. than twenty registered trials employing stem cells from epithelial
tissues on the public clinical trials database (www.clinicaltrials.
4.1. Mesenchymal stem cells gov). The application of epithelial stem cells to regenerative med-
icine differs from MSC as they do not home to sites of inflammation
On the public clinical trials database (www.clinicaltrials.gov) and act in an immunomodulatory manner, nor have they been
there are currently over 350 registered clinical trials utilising MSC- shown to exhibit the same paracrine effects to restore tissue
based cell therapies. MSCs have shown particular promise in the function. Thus therapies involving epithelial stem cells are largely
treatment of tissue injury and degenerative diseases. One of the aimed at either the generation of epithelial tissue in vitro for future
main areas of interest has been in myocardial repair, where MSC transplant into patients, or direct engraftment of stem cells
therapy has been shown to improve cardiac function [29,30], following in vivo administration in order to regenerate damaged
although the exact mechanism by which they are able to do this tissue or replace genetically defective cell populations.
remains controversial. After reaching the heart, MSCs may incor- Skin has been at the forefront of regenerative medicine using
porate into and contribute to the specific functions of cardiac tis- epithelial stem cells. Stratified epithelial stem cells were first used in
sue; this is known as engraftment. However, whilst MSCs are regenerative medicine in the early 1980’s for the treatment of burn
capable of persisting in different tissues for several months patients [48], and subsequent advances and refinements in such
following systemic administration, the proportion of cells that therapies now improve the lives of severe burn patients worldwide
actually engraft is relatively small [31]. Interestingly, injury appears [22]. However, such ex vivo organ culture and transplant is not an
to improve engraftment of MSC and following systemic delivery option applicable to in utero treatment of placental disorders. In vivo
MSC engraftment in ischaemic cardiac tissue is significantly higher regeneration of epithelial tissues has also been undertaken in a
J.L. James et al. / Placenta 35 (2014) 77e84 81

Fig. 2. Schematic diagram demonstrating how MSC modulate the activities of a range of immune cells populations.

number of models, the most successful of which is the cornea of the 5.1. Practical applications
eye. The cornea consists of a transparent flat epithelial layer that is
formed from a specific population of epithelial stem cells, termed As so little is known about the exact role of human TSCs in
limbal stem cells, which reside in the corneal limbus, a narrow zone pregnancy disorders, the most obvious therapeutic applications of
found at the border of the cornea and the sclera (the white of the stem cells to pregnancy disorders at present revolve around MSCs.
eye). Corneal transplants involve allogenic stromal engraftment, An important therapeutic effect of MSCs is the induction of a pro-
after which the outer surface of the cornea is eventually replaced by angiogenic environment. Insufficient and disordered placental
host-derived epithelium generated by undamaged autologous angiogenesis is a common feature of IUGR placentae [52]. In such
limbal stem cells [49]. However, if the limbus is damaged, the cases re-establishment of an efficient blood supply that maximises
epithelium is derived from invasion of conjunctival cells, resulting nutrient exchange is crucial for the fetus to reach its optimal
in vascularisation, inflammation and corneal opacification. growth potential. As MSCs are thought to play a normal physio-
Engraftment of limbal stem cells is able to overcome this problem, logical role in placental angiogenesis, MSCs transplanted into IUGR
and has been shown to restore corneal integrity and visual acuity in placentae may preferentially migrate to sites of oxygen deficit
78% of patients over the past decade of use [49]. within the placenta where they may act on placental blood vessels
As in the cornea, a defining characteristic of epithelial cells to stimulate angiogenesis and improve nutrient transfer and fetal
across a range of tissues is their close apposition to the underlying growth.
mesenchymal cells. Due to the close proximity of these cells it is A number of pregnancy complications also involve placental
perhaps not surprising that interactions between them are a key lesions that damage the vascular tree and impair nutrient transfer,
part of tissue morphogenesis. Mesenchymal cells are able to control and stem cell therapies aimed at tissue regeneration may be
epithelial cell-fate in a number of tissues [50], and in turn epithelial particularly effective in the treatment of these disorders. Placental
cells have been shown to ‘talk back’ to MSCs to facilitate the infarction affects around 30% of hypertensive pregnancies [53],
organisation of tissue structure [22,51]. In a similar manner, the resulting in the interruption of fetal blood supply to a region of the
importance of interactions between MSCs and TSCs throughout placenta and subsequent ischaemic tissue damage [54]. Such in-
pregnancy means that transplanting MSC into the placenta may farcts can exacerbate placental insufficiency and further contribute
also help promote trophoblast function, highlighting the need to to the development of IUGR. In animal models of myocardial
better understand how these cells interact to result in successful infarction, transplantation of placental MSCs results in increased
placentation. blood flow, increased vessel density and delayed necrosis, sug-
gesting that the transplanted MSCs are responsible for inducing
5. Stem cells in the obstetric clinic e can we fix pregnancy angiogenesis [55,56]. By extrapolation, administration of MSCs to
disorders? an infarcted placenta may result in enhanced angiogenesis and
regeneration of the villous tissue.
Unlike most organs, the placenta undergoes extremely rapid In all pre-clinical and clinical studies using MSCs, migration into
growth and differentiation throughout its entire lifespan until its the damaged tissue is a crucial process for the efficacy of treatment.
‘death’ at the time of delivery. This is perhaps why it has shown Therefore, understanding the tissue and cell specific factors that
such promise as a rich source of MSCs, and highlights the oppor- regulate MSC trafficking, both from the site of administration and
tunity that stem cells have to make significant impact on placental to/from their stem cell niche within the placenta, will be crucial for
function. effective administration of MSCs. In vivo, MSCs have been delivered
82 J.L. James et al. / Placenta 35 (2014) 77e84

by both intravenous injection and local intra-arterial injection, with transplantation may be the more effective treatment option.
the former demonstrating that circulating MSCs can specifically Further understanding of TSC function in the future will help
mobilise to sites of injury or inflammation, while the later can elucidate the potential for these cells to be targeted therapeutically,
enhance the accumulation in specific target organs [32,57]. The with potential implications for the function of downstream mature
ability of MSCs to be delivered through an arterial route suggests trophoblast lineages in the placenta.
that they could be administered to the placenta in utero via the
umbilical artery, a delivery method previously established for fetal 5.3. Safety
transfusions in rhesus disease [58]. Such a route would be more
favourable than maternal systemic administration in which a sig- The mantra of ‘first do no harm’ is never more true than when
nificant maternal ‘first pass’ effect is likely to occur whereby stem treating pregnancy disorders, and such caution around adverse
cells would traffic into maternal tissue prior to reaching the uterine fetal outcomes means that the field of obstetric medicine has
circulation, leaving a potentially small proportion of cells available rightly been extremely cautious in developing new therapies.
to cross into the placenta. Therefore, one of the major questions that must be addressed in
considering stem cell based therapies for the treatment of placental
5.2. Allogenic transplantation or therapeutic targeting? disorders is the potential risks involved. Important precedents to
evaluate the potential efficacy and safety of such therapies may
Whilst the isolation of MSCs from fetal cord blood in the third come from the field of fetal medicine, where allogenic intra-uterine
trimester may be an option for autologous treatment of pregnancy transplantation of haematopoietic and neural stem cell populations
disorders, due to the relatively short window of treatment limiting has been trialled in a number of animal models in order to develop
the time these cells could be expanded in culture, and the potential treatments for a range of lymphohematopoietic, neural, metabolic
for autologous MSCs to suffer from the same underlying defects as and genetic disorders [64]. Indeed, successful treatment of osteo-
those that resulted in inadequate placental function in the first place, genesis imperfecta (a degenerative bone disorder) was achieved by
allogenic transplantation of stem cells is likely to be necessary to treat the injection of allogeneic MSC into the umbilical vessels at 32
placental disorders. Fortunately due to their unique immunopheno- weeks of gestation, with no signs of fetal distress and successful
type, allogenically transplanted MSC are well tolerated. A recent preservation of the pregnancy [65].
clinical trial comparing the effects of allogeneic and autologous MSC Treatments for placental disorders would clearly need to be
in patients with ischaemic cardiac damage found that improvements extensively validated, first in in vitro models to understand how
in cardiac tissue structure were comparable between the two groups, stem cells injected into the placenta directly or via the umbilical
and only 3.7% of patients generated donor-specific antibodies in the circulation may migrate and function within the tissue, and then in
absence of immune suppression [30]. The placenta is the only natu- haemochorial animal models in order to evaluate not only their
rally occurring tissue transplant and consequently placental MSCs safety and efficacy, but the potential for fetal chimerism as a result of
may induce an even greater level of immunotolerance than those cellular engraftment outside the placenta, and the long term effects
derived from other tissue types due to their lack of expression of that this may have. However, the more we understand about stem
major histocompatibility class-II (MHC II) and the co-stimulatory cell therapies in adult regenerative medicine, the more we may be
molecules (CD80, CD86, CD83 and CD40) [19,18]. able to circumvent any potential issues in intra-uterine stem cell
When considering the situation in utero it is unlikely the pri- delivery by reducing engraftment, priming stem cells for specific
mary host e the fetus itself - would reject the transplanted cells, as tasks, rendering cells non-proliferative prior to administration, or
its immune system is relatively immature and still learning to alternatively developing pharmaceutical agents that specifically
recognise self from non-self antigens. Indeed, allogenic haemato- target stem cells within the placenta to correct deficiencies in their
poietic stem cell transplantation in utero is able to induce donor function or induce proliferation and/or differentiation.
specific tolerance in mice [59]. However, a secondary host must
also be considered e the mother. The placental barrier is somewhat 6. Conclusions
permeable, with maternal leukocytes able to traffic into the fetus in
response to intra-uterine transplantation of haematopoietic stem So the question remains e can we fix it? The significant promise
cells in mice [60], and fetal cells, including MSC, released from the that stem cell therapies have for the clinical treatment of pregnancy
placenta may also engraft in maternal tissues resulting in maternal disorders means that surely we must try. Both MSC and TSC have
microchimerism [61], thus the ability of allogenically transplanted therapeutic potential, but the differences in function of these stem
placental stem cells to successfully hide from the maternal immune cell types mean that while MSC may be able to be directly trans-
system is imperative to the success of such treatments. planted into the placenta and migrate to sites of action, placental
If allogenic transplantation does not prove to be therapeutically inadequacies arising from potential TSC dysfunction may be better
viable, an alternative approach could be to stimulate stem cell treated by exogenously stimulating stem cells already resident in
function pharmacologically using peptides or nanoparticles specific the placenta. Many fields of medicine have advanced by adminis-
to placental stem cell populations [62,63]. This may be of particular tering treatments that have shown to be effective and safe without
importance for the therapeutic targeting of TSC as it is unclear fully understanding the mechanisms of action behind their efficacy.
whether epithelial stem cells transplanted outside their niche are However, due to the plasticity of the fetus in utero, in obstetrics we
able to migrate within tissue to sites of action. The organised must take the more measured approach and obtain a comprehen-
manner in which the migration of epithelial stem cells away from sive understanding of how stem cells function in both healthy and
their niche regulates their differentiation programme, combined dysfunctional placentae, before we can determine how best to tap
with the fact that epithelial stem cells do not circulate in the blood into their therapeutic potential to treat pregnancy disorders.
like MSC, and are not found outside epithelial tissues, suggests that
their ability to function as stem cells is much more niche dependent Acknowledgements
than MSC. Therefore epithelial stem cells are unlikely to be suitable
targets for intravenous injection, and in organs such as the placenta Dr James is supported by a grant from the Auckland Medical
where site-specific transplantation is unlikely to be feasible, phar- Research Foundation. The authors wish to thank Dr Olivia Holland
macological stimulation of these cells rather than direct for allowing the use of her image of a first trimester placenta.
J.L. James et al. / Placenta 35 (2014) 77e84 83

References [27] Ligi I, Simoncini S, Tellier E, Vassallo PF, Sabatier F, Guillet B, et al. A switch
toward angiostatic gene expression impairs the angiogenic properties of
endothelial progenitor cells in low birth weight preterm infants. Blood
[1] James JL, Carter AM, Chamley LW. Human placentation from nidation to 5
2011;118(6):1699e709.
weeks of gestation. Part I: what do we know about formative placental
[28] Demir R, Kayisli UA, Cayli S, Huppertz B. Sequential steps during vasculo-
development following implantation? Placenta 2012;33(5):327e34.
genesis and angiogenesis in the very early human placenta. Placenta
[2] Boyd J, Hamilton W. The human placenta. Cambridge: W. Heffer & Sons Ltd;
2006;27(6e7):535e9.
1970.
[29] Przybyt E, Harmsen MC. Mesenchymal stem cells: promising for myocardial
[3] Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF,
regeneration? Curr Stem Cell Res Ther 2013;8(4):270e7.
Luria EA, et al. Precursors for fibroblasts in different populations of hemato-
[30] Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP,
poietic cells as detected by the in vitro colony assay method. Exp Hematol
Suncion VY, et al. Comparison of allogeneic vs autologous bone marrow-
1974;2(2):83e92.
derived mesenchymal stem cells delivered by transendocardial injection in
[4] Castrechini NM, Murthi P, Gude NM, Erwich JJ, Gronthos S, Zannettino A, et al.
patients with ischemic cardiomyopathy: the POSEIDON randomized trial.
Mesenchymal stem cells in human placental chorionic villi reside in a vascular
JAMA 2012;308(22):2369e79.
Niche. Placenta 2010;31(3):203e12.
[31] Devine SM, Cobbs C, Jennings M, Bartholomew A, Hoffman R. Mesenchymal
[5] Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new
stem cells distribute to a wide range of tissues following systemic infusion
trend for cell therapy. Acta Pharmacol Sin 2013;34(6):747e54.
into nonhuman primates. Blood 2003;101(8):2999e3001.
[6] Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D,
[32] Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, et al.
et al. Minimal criteria for defining multipotent mesenchymal stromal cells.
Systemic delivery of bone marrow-derived mesenchymal stem cells to the
The International Society for Cellular Therapy position statement. Cytotherapy
infarcted myocardium: feasibility, cell migration, and body distribution. Cir-
2006;8(4):315e7.
culation 2003;108(7):863e8.
[7] Demir R, Kayisli UA, Seval Y, Celik-Ozenci C, Korgun ET, Demir-Weusten AY,
[33] von Bahr L, Batsis I, Moll G, Hagg M, Szakos A, Sundberg B, et al. Analysis of
et al. Sequential expression of VEGF and its receptors in human placental villi
tissues following mesenchymal stromal cell therapy in humans indicates
during very early pregnancy: differences between placental vasculogenesis
limited long-term engraftment and no ectopic tissue formation. Stem Cells
and angiogenesis. Placenta 2004;25(6):560e72.
2012;30(7):1575e8.
[8] Demir R, Kaufmann P, Castellucci M, Erbengi T, Kotowski A. Fetal vasculo-
[34] Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, et al. Local delivery
genesis and angiogenesis in human placental villi. Acta Anat (Basel)
of marrow-derived stromal cells augments collateral perfusion through
1989;136(3):190e203.
paracrine mechanisms. Circulation 2004;109(12):1543e9.
[9] Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, et al. Bone
[35] Hung SC, Pochampally RR, Chen SC, Hsu SC, Prockop DJ. Angiogenic effects of
marrow origin of endothelial progenitor cells responsible for postnatal vas-
human multipotent stromal cell conditioned medium activate the PI3K-Akt
culogenesis in physiological and pathological neovascularization. Circ Res
pathway in hypoxic endothelial cells to inhibit apoptosis, increase survival,
1999;85(3):221e8.
and stimulate angiogenesis. Stem Cells 2007;25(9):2363e70.
[10] Meraviglia V, Vecellio M, Grasselli A, Baccarin M, Farsetti A, Capogrossi MC,
[36] Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, Silberstein LE.
et al. Human chorionic villus mesenchymal stromal cells reveal strong
Human bone marrow stromal cells express a distinct set of biologically
endothelial conversion properties. Differentiation 2012;83(5):260e70.
functional chemokine receptors. Stem Cells 2006;24(4):1030e41.
[11] Warrier S, Haridas N, Bhonde R. Inherent propensity of amnion-derived
[37] Ringe J, Strassburg S, Neumann K, Endres M, Notter M, Burmester GR, et al.
mesenchymal stem cells towards endothelial lineage: vascularization from
Towards in situ tissue repair: human mesenchymal stem cells express che-
an avascular tissue. Placenta 2012;33(10):850e8.
mokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation
[12] Tremain N, Korkko J, Ibberson D, Kopen GC, DiGirolamo C, Phinney DG.
with CXCL8 but not CCL2. J Cell Biochem 2007;101(1):135e46.
MicroSAGE analysis of 2,353 expressed genes in a single cell-derived colony of
[38] Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of
undifferentiated human mesenchymal stem cells reveals mRNAs of multiple
allogeneic T-cell proliferation by human marrow stromal cells: implications in
cell lineages. Stem Cells 2001;19(5):408e18.
transplantation. Transplantation 2003;75(3):389e97.
[13] Lash GE, Naruse K, Innes BA, Robson SC, Searle RF, Bulmer JN. Secretion of
[39] Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using
angiogenic growth factors by villous cytotrophoblast and extravillous
allogeneic bone marrow mesenchymal stem cells prevents tissue damage in
trophoblast in early human pregnancy. Placenta 2010;31(6):545e8.
collagen-induced arthritis. Arthritis Rheum 2007;56(4):1175e86.
[14] Gang EJ, Jeong JA, Han S, Yan Q, Jeon CJ, Kim H. In vitro endothelial potential of
[40] Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate alloge-
human UC blood-derived mesenchymal stem cells. Cytotherapy 2006;8(3):
neic immune cell responses. Blood 2005;105(4):1815e22.
215e27.
[41] Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L.
[15] Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G, Bornhauser M,
Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity,
et al. Mesenchymal stem cells can be differentiated into endothelial cells
and cytokine production: role of indoleamine 2,3-dioxygenase and prosta-
in vitro. Stem Cells 2004;22(3):377e84.
glandin E2. Blood 2008;111(3):1327e33.
[16] Sung HJ, Hong SC, Yoo JH, Oh JH, Shin HJ, Choi IY, et al. Stemness evaluation of
[42] Abumaree MH, Al Jumah MA, Kalionis B, Jawdat D, Al Khaldi A, Abomaray FM,
mesenchymal stem cells from placentas according to developmental stage:
et al. Human placental mesenchymal stem cells (pMSCs) play a role as im-
comparison to those from adult bone marrow. J Korean Med Sci 2010;25(10):
mune suppressive cells by shifting macrophage differentiation from inflam-
1418e26.
matory M1 to anti-inflammatory M2 macrophages. Stem Cell Rev 2013;9(5):
[17] Benirschke K, Kaufmann P. Pathology of the human placenta. New York:
620e41.
Springer-Verlag; 2000.
[43] Shohara R, Yamamoto A, Takikawa S, Iwase A, Hibi H, Kikkawa F, et al.
[18] James JL, Carter AM, Chamley LW. Human placentation from nidation to 5
Mesenchymal stromal cells of human umbilical cord Wharton’s jelly accel-
weeks of gestation. Part II: tools to model the crucial first days. Placenta
erate wound healing by paracrine mechanisms. Cytotherapy 2012;14(10):
2012;33(5):335e42.
1171e81.
[19] James J, Stone P, Chamley L. The isolation and characterization of a population
[44] Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell
of extravillous trophoblast progenitors from first trimester human placenta.
signaling and therapy. Circ Res 2008;103(11):1204e19.
Hum Reprod 2007;22(8):2111e9.
[45] Mirotsou M, Zhang Z, Deb A, Zhang L, Gnecchi M, Noiseux N, et al. Secreted
[20] Telugu BP, Adachi K, Schlitt JM, Ezashi T, Schust DJ, Roberts RM, et al. Com-
frizzled related protein 2 (Sfrp2) is the key Akt-mesenchymal stem cell-
parison of extravillous trophoblast cells derived from human embryonic stem
released paracrine factor mediating myocardial survival and repair. Proc
cells and from first trimester human placentas. Placenta 2013;37(7):536e43.
Natl Acad Sci U S A 2007;104(5):1643e8.
[21] Aboagye-Mathiesen G, Laugesen J, Zdravkovic M, Ebbesen P. Isolation and
[46] Chen CP, Lee MY, Huang JP, Aplin JD, Wu YH, Hu CS, et al. Trafficking of
characterization of human placental trophoblast subpopulations from first
multipotent mesenchymal stromal cells from maternal circulation through
trimester chorionic villi. Clin Diagn Lab Immunol 1996;3:14e22.
the placenta involves vascular endothelial growth factor receptor-1 and
[22] Blanpain C, Horsley V, Fuchs E. Epithelial stem cells: turning over new leaves.
integrins. Stem Cells 2008;26(2):550e61.
Cell 2007;128(3):445e58.
[47] Xiang MX, He AN, Wang JA, Gui C. Protective paracrine effect of mesenchymal
[23] Rolfo A, Giuffrida D, Nuzzo AM, Pierobon D, Cardaropoli S, Piccoli E, et al. Pro-
stem cells on cardiomyocytes. J Zhejiang Univ Sci B 2009;10(8):619e24.
inflammatory profile of preeclamptic placental mesenchymal stromal cells:
[48] Grafting of burns with cultured epithelium prepared from autologous
new insights into the etiopathogenesis of preeclampsia. PLoS One 2013;8(3):
epidermal cells. Lancet 1981;1(8211):75e8.
e59403.
[49] Pellegrini G, Rama P, Mavilio F, De Luca M. Epithelial stem cells in corneal
[24] Hwang JH, Lee MJ, Seok OS, Paek YC, Cho GJ, Seol HJ, et al. Cytokine expression
regeneration and epidermal gene therapy. J Pathol 2009;217(2):217e28.
in placenta-derived mesenchymal stem cells in patients with pre-eclampsia
[50] Birchmeier C, Birchmeier W. Molecular aspects of mesenchymal-epithelial
and normal pregnancies. Cytokine 2010;49(1):95e101.
interactions. Annu Rev Cell Biol 1993;9:511e40.
[25] Wang Y, Fan H, Zhao G, Liu D, Du L, Wang Z, et al. miR-16 inhibits the pro-
[51] Hardy MH. The secret life of the hair follicle. Trends Genet 1992;8(2):55e61.
liferation and angiogenesis-regulating potential of mesenchymal stem cells in
[52] Clausson B, Gardosi J, Francis A, Cnattingius S. Perinatal outcome in SGA births
severe pre-eclampsia. FEBS J 2012;279(24):4510e24.
defined by customised versus population-based birthweight standards. BJOG
[26] Sipos PI, Bourque SL, Hubel CA, Baker PN, Sibley CP, Davidge ST, et al. Endo-
2001;108(8):830e4.
thelial colony forming cells derived from pregnancies complicated by intra-
[53] Salgado SS, Pathmeswaran A. Effects of placental infarctions on the fetal
uterine growth restriction are fewer and have reduced vasculogenic capacity.
outcome in pregnancies complicated by hypertension. J Coll Physicians Surg
J Clin Endocrinol Metab 2013;98(12):4953e60.
Pak 2008;18(4):213e6.
84 J.L. James et al. / Placenta 35 (2014) 77e84

[54] McDermott M, Gillan JE. Chronic reduction in fetal blood flow is associated [60] Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q, MacKenzie TC. Maternal T
with placental infarction. Placenta 1995;16(2):165e70. cells limit engraftment after in utero hematopoietic cell transplantation in
[55] Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates mice. J Clin Invest 2011;121(2):582e92.
wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. [61] Pritchard S, Wick HC, Slonim DK, Johnson KL, Bianchi DW. Comprehensive
Biochem Biophys Res Commun 2013;438(2):410e9. analysis of genes expressed by rare microchimeric fetal cells in the maternal
[56] Tran TC, Kimura K, Nagano M, Yamashita T, Ohneda K, Sugimori H, et al. mouse lung. Biol Reprod 2012;87(2):42.
Identification of human placenta-derived mesenchymal stem cells involved in [62] Harris LK, Kotamraju VR, Teesalu T, Ruoslahti E. Identification of novel
re-endothelialization. J Cell Physiol 2011;226(1):224e35. placental homing peptides. Placenta 2012;33(9):A44.
[57] Freyman T, Polin G, Osman H, Crary J, Lu M, Cheng L, et al. A quantitative, [63] Jones H, Klanke C, Ayres N, Habli M, Shaaban A, Keswani S, et al. Nanoparticle-
randomized study evaluating three methods of mesenchymal stem cell de- mediated trophoblast-specific gene transfer in an in vitro model of human
livery following myocardial infarction. Eur Heart J 2006;27(9):1114e22. trophoblast. Reprod Sci 2013;20(3):303A.
[58] Schumacher B, Moise Jr KJ. Fetal transfusion for red blood cell alloimmuni- [64] Fauza D. Tissue engineering and transplantation in the fetus. Elsevier; 2014.
zation in pregnancy. Obstet Gynecol 1996;88(1):137e50. pp. 511e29.
[59] Ashizuka S, Peranteau WH, Hayashi S, Flake AW. Busulfan-conditioned bone [65] Le Blanc K, Götherström C, Ringdén O, Hassan M, McMahon R, Horwitz E, et al.
marrow transplantation results in high-level allogeneic chimerism in mice Fetal mesenchymal stem-cell engraftment in bone after in utero trans-
made tolerant by in utero hematopoietic cell transplantation. Exp Hematol plantation in a patient with severe osteogenesis imperfecta. Transplantation
2006;34(3):359e68. 2005;79(11):1607e14.

S-ar putea să vă placă și