Anda di halaman 1dari 6

Placenta 34, Supplement A, Trophoblast Research, Vol.

27 (2013) S62eS67

Contents lists available at SciVerse ScienceDirect

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

Review: Endothelial progenitor cells in pregnancy and obstetric


pathologies
I.P. Crocker a, b, *, P.I. Sipos a, b
a
Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, UK
b
Maternal and Fetal Health Research Centre, St. Mary’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic
Health Science Centre, Manchester M13 9WL, UK

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

Article history: Since their discovery, endothelial progenitor cells (EPCs) have generated considerable interest in vascular
Accepted 3 January 2013 biology. They are a heterogeneous population of cells that exist in both the fetus and adult, and are
mobilized to support de novo vessel formation or encourage vascular health. This review summarizes our
Keywords: understanding of these cells in pregnancy, paying particular attention to their physiological role in
Endothelial progenitor cells placental development and the uterus, alongside their involvement in related obstetric pathologies.
Placenta
Ó 2013 Published by IFPA and Elsevier Ltd.
Uterus
Pre-eclampsia
Intrauterine growth restriction

1. Introduction Since 1997, the number of investigations into human EPCs has
grown exponentially. The bulk of this research has focused on adult
Vasculogenesis and angiogenesis are processes of new blood cardiovascular disease, with scant attention to the fetus, placenta or
vessel generation. Before 1997, vasculogenesis, de novo vessel for- pregnancy. The fact that vessel formation and vascular expansion
mation through differentiation of angioblasts, was believed to be are implicit to pregnancy, and that the presence and activity of these
restricted to embryonic development, with angiogenesis, forma- cells is greater in the fetus compared to adults, has generally gone
tion from pre-existing vessels, superseding postnatally. In 1997 unnoticed. This article reviews current evidence of EPC involvement
Asahara et al. [1] published the first report of circulating bone- in human pregnancy. It appraises the current literature, introduces
marrow derived cells in humans capable of differentiation into some preliminary findings and discusses possibilities for further
mature endothelia and participation in neovascularisation. Shi obstetric research and intervention. The involvement of EPCs in the
et al., then showed comparable cells endothelialising intra-aortic pathophysiology of pre-eclampsia, intrauterine growth restriction
Dracon grafts in dogs [2]. These two studies established the exis- (IUGR) and fetal programming is considered.
tence of endothelial progenitor cells (EPCs) in the adult, with
functional similarities to embryonic angioblasts. With this discov- 2. Origins of EPCs
ery came a paradigm shift in vascular biology, with the recognition
that neovascularisation in adults could depend, at least in part, on In recent years EPCs, and more specifically their absence, have
putative postnatal vasculogenesis. Subsequent studies confirmed emerged as risk factors for cardiovascular disease, with good evi-
that EPCs are mobilized from the bone-marrow (and possibly from dence for a role in endothelial homeostasis and repair. Independent
other organs) and home to sites of tissue ischemia or endothelial of more classic risk factors, the levels of these cells can predict
damage, adopting a more mature endothelial phenotype and con- endothelial dysfunction in healthy individuals, and cardiovascular
tributing to de novo vessel formation. Thus, EPCs play an integral events in patients with coronary artery disease [3,4]. A similar
role in endothelium regeneration, vessel repair and maintenance. reduction in circulating EPCs is noted in a variety of other vascular-
related conditions, including diabetes [5], peripheral vascular dis-
ease, rheumatoid arthritis [6] and obesity [7], but the mechanisms
* Corresponding author. Maternal and Fetal Health Research Centre, Institute of underlying these associations are uncertain.
Human Development, Faculty of Medical and Human Sciences, University of
Manchester, Reception, 5th Floor (Research), St Mary’s Hospital, Oxford Road,
Although not restricted to bone marrow, the majority of EPCs
Manchester, M13 9WL, UK. Tel.: þ44 (0) 161 701 6973. reside within its microenvironment, termed the stromal or osteo-
E-mail address: Ian.Crocker@manchester.ac.uk (I.P. Crocker). blast niche. In this state they are tethered by integrins to the stromal

0143-4004/$ e see front matter Ó 2013 Published by IFPA and Elsevier Ltd.
http://dx.doi.org/10.1016/j.placenta.2013.01.003
I.P. Crocker, P.I. Sipos / Placenta 34, Supplement A, Trophoblast Research, Vol. 27 (2013) S62eS67 S63

cells until they are transferred to the vascular niche for subsequent culture, are highly proliferative, can be clonally expanded, but are
egress to the peripheral circulation. All classes of EPCs are derived otherwise indistinguishable from mature endothelial cells. CACS and
from hemangioblasts, embryonic or adult, which differentiate to ECFCs may work synergistically in vivo, CACs primarily enhancing the
endothelial cells or myeloid progenitors (Fig. 1). Ultimately, whether survival and function of ECFCs by providing copious angiogenic fac-
all endothelial cell progenitors proceed through a “monocytic” phase tors (NO, VEGF, IGF-I etc), and ECFCs participating in blood vessel and
is unclear. However, at least two fundamentally different types of endothelium formation per se [9]. Flow cytometry is also used to
EPCs may be mobilized, CD45 and CD34 expressing hematopoietic- enumerate these groups. These approaches most often use CD34 or
like cells and CD34 positive CD45 negative “endothelial-like” CD133 in combination with VEGFR-2 (KDR), but again a consensus is
equivalents [8]. These cells play a critical and some say comple- far from reached. In general, ECFCs are often characterized as CD31þ/
mentary role in blood vessel formation and repair [9]. CD34þ/CD45/VEGFR-2þ/CD133, CACs as CD31þ/CD34þ/CD45þ/
The proliferation and mobilization of EPCs from the bone-marrow dim
/CD14þ/CD133þ/ [13]. These techniques are improved by
involve a complex interplay of adhesion molecules, chemokines, excluding false positive events, such as dead cells and CD3þ T cells.
cytokines and proteinases. In vascular injury and tissue ischemia,
vascular endothelial growth factor (VEGF) and stromal cell-derived
4. EPCs and the uterus
factor-1 (SDF-1) are thought to be particularly important, upregu-
lated in the circulation by hypoxia-inducible factor 1 (HIF-1) [10].
Extensive remodeling and growth within the human endome-
Nitric oxide (NO) is also a key intermediary, as the activation of
trium precedes implantation. Physiological angiogenesis is funda-
endothelial nitric oxide synthase (eNOS) in the bone marrow is
mental to the proliferative and secretory phases of the menstrual
considered a common mobilizing reaction of EPCs to VEGF, estrogen,
cycle. The involvement of adult stem cells in the cyclic regeneration
insulin-like growth factor I (IGF-I) and erythropoietin [11]. In lib-
and shedding of the endometrium was first suggested by Chan et al.,
erating EPCs, SDF-1 and NO activate matrix metalloproteinase 9
in 2004, following the clonogenic derivation of human endometrial
(MMP-9), which in turn catalyzes the proteolysis of Kit Ligand (KitL),
epithelial and stromal cells [14]. Although, EPCs had already been
severing its adhesion to c-kit receptor and freeing the cells from their
defined in the endometrium in the mouse at this time, their role had
bone marrow niche. In mice, acute administration of this receptor, in
not been confirmed [15]. In 2007, bone-marrow derived EPCs were
its soluble form, sc-kit, increases mobilization of EPCs, theoretically
shown to contribute to de novo blood vessel formation in the mouse
disrupting these c-kit - KitL interactions [12].
endometrium [16]. In verifying EPC participation in the menstrual
cycle, flow cytometric determinations of circulating human cells
3. Characterization of EPCs were performed. Although not definitive, these studies showed el-
evations in the secretory and follicular phases, implying steroid
The characterization of EPCs is highly contentious. In cell cultures regulation and endometrial regenerative contributions [17,18].
of unfractionated mononuclear cells, at least two fundamentally Given that local factors such as estradiol, tumor necrosis factor-
different types of EPCs have been generated, early- and late- a (TNF-a), interleukin (IL)-6, VEGF and intercellular adhesion mol-
outgrowth cells, respectively. The early outgrowth cells are referred ecule (ICAM)-1, participate in EPC migration and trafficking, their
to as colony-forming unit endothelial cells (CFU-ECs) or circulating contribution to the human cycle was anticipated, but sex steroids
angiogenic cells (CACs), whilst alternative conditions yield endo- and circulating inflammatory mediators have yet to be correlated
thelial colony forming cells (ECFCs) (Fig. 1). These cells appear late in with peripheral EPCs in the non-pregnant state [17].

Fig. 1. Pathways to EPC ontogeny. Primitive haemangioblasts in the bone marrow give rise to myeloid and endothelial progenitors which yield early outgrowth, ‘endothelial-like’
colony-forming unit cells (ECFCs) or spindle-shaped circulating angiogenic cells (CACs). The relative expression of cell surface markers identifies these differentiation pathways.
ECFCs eventually form mature endothelium, whilst CACs orchestrate neovascularisation.
S64 I.P. Crocker, P.I. Sipos / Placenta 34, Supplement A, Trophoblast Research, Vol. 27 (2013) S62eS67

Throughout pregnancy, the uterus undergoes widespread vas- 7. EPCs and placental pathologies
culature adaptations. In addition to extensive vasodilatation and
anastomosis [19], uterine spiral arteries undergo elaborate Within the human placenta, angiogenic activities are orchestrated
remodeling, mediated by interstitial and endovascular trophoblast. by numerous factors, including oxygen tension and pro-and anti-
During trophoblast invasion, maternal spiral arteries are trans- angiogenic mediators, such as placental growth factor (PlGF), VEGF
formed into wide-bore conduits, in which the endothelia are first and its neutralizing soluble receptor sVEGFR-1. With studies in pre-
damaged and then replenished. Although trans-differentiation of eclampsia indicating variations of such molecules at the placental
trophoblast was initially conceived to replace these losses, current level [28], their impact on fetal endothelial cells has also been con-
evidence supports re-endothelialisation [20]. In preliminary stud- sidered. In vitro, and thus in the absence of these factors, isolated
ies, we have investigated the involvement of fetal EPCs in these HUVECs, i.e. mature endothelial cells, from preeclamptic pregnancies
uterine adaptive events [21]. This concept was derived in the have shown inherent alterations in cell permeability, morphology,
knowledge that adult EPC efficiency is dramatically surpassed by mechanical properties, release of vasodilatory mediators and
their fetal counterparts [22]. In mouse models, we tentatively enhanced angiogenic capacity [29e32]. Although their significance is
concluded that fetal ECFCs transmigrate into the maternal blood- unproven, these anomalies are attributed to intrinsic differences or
stream and home to locations of maternal vasculogenesis, primarily environmental adaptations prior to cell isolation, i.e. the angiogenic
microvessels of the pregnant uterus [21]. If verified, this chimerism feto-placental milieu. In similar experiments, cord-derived EPCs from
offers the tantalizing involvement of fetal EPCs in human uterine women with severe pre-eclampsia have shown a reduction in num-
adaptations, and provides potential alternative participants in bers and attenuated proliferation, migration and vasculogenic ca-
spiral artery endothelium restoration. pacity [33,34]. These anomalies are specifically related to diminished
cord plasma free VEGF and overabundance of sVEGFR-1 (sFlt-1) [35].
In line with pre-eclampsia studies, attenuated changes in cord
5. EPCs and endometriosis
blood VEGF and sFlt-1 have also been recorded in cases of IUGR, i.e.
pregnancies more associated with abnormal placental angiogenesis
In addition to the recruitment of EPCs into the human endo-
and feto-placental vascular anomalies [36]. Our initial studies of
metrium, either in pregnancy or the menstrual cycle, the possibility
cord blood in IUGR have considered EPC availability and in vitro
that these cells are liberated in retrograde fashion is also enter-
activities [37]. In comparison with uncomplicated pregnancies,
tained. As circulating endothelial cells are shed artificially or
provisional flow cytometry data, suggested that fewer EPCs (CACs
spontaneously from vessels, particularly during trauma, it is con-
and ECFCs) were present in growth restricted pregnancies and that
ceivable that endometrial EPCs slough off from endometrial vessels
placental sequestration was also impaired [37]. The culture prop-
and are mobilized to ectopic sites through blood or lymphatic
agation of ECFCs confirmed these findings, with fewer colonies and
systems. In a unifying concept for the pathogenesis of endome-
longer doubling times, hinting at diminished proliferative capacity
triosis, Maruyama and colleagues proposed that these endometrial
[37]. In support, Hwang et al. recently confirmed these prolonged
EPCs migrate into the parenchyma of ectopic organs and differ-
differentiation times and highlighted impaired senescence through
entiate into endometrial cell components, thus giving rise to
diminished telomerase and increased b-galactosidase activity [38].
endometrial lesions [23]. In support, circulating EPCs are upregu-
Moreover, in a related study, Ligi et al. investigated whether low
lated in a mouse model of endometriosis [24], whilst the same
birthweight and prematurity could impact the angiogenic prop-
group recently showed that endometrial stem/progenitor pop-
erties of cord blood ECFCs [39]. As well as deficits in number, and
ulations display significant endothelial-like characteristics, along-
angiogenic defects in vitro and in vivo, these investigators under-
side more traditional endometrial progenitor properties [25].
took gene profiling and highlighted increased expression of anti-
angiogenic genes, predominately thrombospondin 1, as a possible
6. EPCS in the fetus and placenta explanatory factor. From these studies, it remains uncertain
whether prior disturbance in ECFC function results in defective
Although EPCs are recruited from the bone marrow, Ingram placental angiogenesis/vasculogenesis, thus leading to IUGR, or
et al. used single cell clonogenic assays to show that a small per- whether inadequate placentation might occur for other reasons,
centage of human umbilical vein endothelial cells (HUVEC) and but with equal and lasting effects on ECFC numbers and function.
human aortic endothelial cells contain endothelial colony-forming Regardless, these studies offer new perspectives on placental pa-
cells of low proliferative potential (LPP-ECFCs), generating colonies thologies, and interesting novel pathways to fetal deprogramming.
with less than 500 cells, and high proliferative potential (HPP-
ECFCs), with improved telomerase activity [22]. In head-to-head 8. Circulating EPCs in pregnancy
comparisons, it was clear not only that these fetal EPCs were
more abundant and active than their adult counterparts, but that Human pregnancy requires sustained cardiovascular adapta-
this high proliferative phenotype was absent in adults altogether tions, typified by a fall in peripheral resistance, offsetting elevations
[22]. As several organs, including the placenta, are capable of pro- in cardiac output and plasma volume. To overcome the ‘vascular
ducing hematopoietic cells [26], it is possible that the placenta is stress’ of pregnancy, the maternal endothelium shows enhanced
a primary or supplementary source of EPCs for the fetus. Given the reactivity and an inflated dependence on autocrine/paracrine va-
unique anatomical position and accessibility of the human pla- sodilators, including NO and prostacyclin. In assessing EPCs, cross-
centa, this idea is readily tested. In further preliminary studies [27], sectional studies of circulating cells in uncomplicated pregnancies
we used stringent and accepted markers of ECFCs and CACs in have been published, with the range of methods generally defying
uncomplicated pregnancies, and showed higher levels of both cells direct comparisons. From flow cytometric enumerations, Luppi
in the umbilical arteries than vein, drawing the likely conclusions et al. reported a progressive increase in circulating CD133þ/VEGFR-
that: (i) the human placenta is a doubtful source of EPCs for the 2þ cells from the first trimester onwards, with a significant rise in
fetus, and (ii) that the placenta retains EPCs on passage through the CD34þ/VEGFRþ cells nearer term [40]. Typically their first trimester
organ [27]. Although these observations await confirmation, we values were below detection, similar to those of non-pregnant
tentatively propose that EPCs have a likely vascular role in the controls. In agreement, Buemi et al. reported similar progressive
placenta, even in late gestation when angiogenesis is curtailed. elevations [41]. However, both studies are in stark contrast to
I.P. Crocker, P.I. Sipos / Placenta 34, Supplement A, Trophoblast Research, Vol. 27 (2013) S62eS67 S65

Matsubara and colleagues, who observed a significant decline in attributed to structural alterations in heparan sulfate proteoglycans
CD34þ/CD133þ/KDRþ cells with gestation and a concomitant [53], perhaps tying pregnancy complications to maternal age.
reduction in EPC numbers and restricted responses to TNF-a and Within the fetus, aberrant EPC numbers and function could the-
angiotensin II in culture [42]. oretically impact the embryonic endothelium directly and impair the
Culture generated EPCs are generally considered more prolif- formation of normal placental vessels, thus restricting nutrient
erative and abundant from the second half of pregnancy, with supply and exacerbating fetal growth restriction. As vascular repair is
a predominance of late outgrowth EPC-CFUs correlating with a perquisite for cardiovascular health, it would be predicted that fetal
serum estradiol [43,44]. Given their superior proliferative capacity, EPCs that underperform in the fetus could equally underperform in
a fetal origin was considered. However, two independent studies later life. Epidemiological evidence supports this notion, as babies
refute this idea, failing to detect the SRY gene in EPCs from pregnant affected by IUGR and pre-eclampsia show increased susceptibility to
women carrying solely male offspring [34,45]. As estrogen itself can cardiovascular disease in adulthood. If verified, EPCs could thus offer
mobilize EPCs from the bone marrow in vivo, and stimulate VEGF an explanation for Barker’s hypothesis; i.e. the association between
production and inhibit EPC senescence in vitro, it seems likely that sub-optimal intrauterine growth and adult-onset chronic disease.
hormonal conditioning is a primary means of optimizing EPCs in Undoubtedly further research is warranted.
pregnancy. In rat models of carotid injury, exogenous estradiol can As studies of EPCs move towards diagnostic and therapeutic
accelerate re-endothelialisation via EPC proliferation/mobilization exploitation, a better understanding of the subgroups involved,
[46]. Whilst additional mouse studies confirm the importance of and their interactions with the key vascular beds of pregnancy, will
the estrogen receptor (ER) in mitigating EPC responses to ischemia become a necessity. At this stage it could be envisaged that sig-
[47]. It would therefore seem that in addition to the known vaso- nificant clinical benefit may be derived. This benefit may take
protective affects of estrogens, through NO stimulation and ROS many forms: (i) the assessments of EPC numbers and function
suppression, further protection, in and outside of pregnancy, is could be used to develop diagnostic tools for related pregnancy
afforded by bolstering effects on EPC efficiency. complications, such as IUGR and pre-eclampsia; (ii) medical in-
terventions could be targeted to improve EPC activation in preg-
nancy (see Table 1 for current possibilities), and (iii) EPCs
9. EPCs and pre-eclampsia
themselves could be used as therapeutic tools, either as autologous
or donor therapies for the fetus, placenta or pregnant woman.
Despite advances, the causes of pre-eclampsia remain ill defined.
From a general perspective, the fledging trials of statins in pre-
Nevertheless, it is generally agreed that early onset cases have
eclampsia may inform this area, whilst a recent study by Wang
deficient placentation, typified by decreased spiral artery remod-
et al., has advocated integrin-linked kinase (ILK) as a target for
eling, whilst late onset syndrome is attributed more to maternal
clinical cell-based gene therapy for pre-eclampsia, after their EPC
vascular maladaptations. Vascular endothelial dysfunction precedes
transfections augmented the angiogenic properties of pre-
clinical diagnosis in both scenarios, as indicated by raised soluble
eclampsia-derived cells in vitro [54].
markers and reduced flow-mediated dilatation. For the most part,
predisposing maladies for pre-eclampsia, i.e. age, renal disease and
diabetes, have intrinsically lower EPCs [48e50]. Therefore similar Conflict of interest statement
studies have emerged for pre-eclampsia, but again with conflicting
results. Whilst Mastubara et al., reported no change in circulating We confirm that there are no known conflicts of interest asso-
EPCs in pre-eclampsia, as determined by flow cytometry [42], their ciated with this publication and there has been no significant
mononuclear derived outgrowth cells showed enhanced prolifera- financial support for this work that could have influenced its
tion upon TNF-a and angiotensin II stimulation. This finding con- outcome.
trasts with that of Sugawara and colleagues, who reported
a decrease in the number of circulating EPCs in pre-eclampsia as
defined by EPC-CFU count e a general correlate of cardiovascular Table 1
risk e and higher rates of senescence [51]. This latter study agrees Pharmaceuticals known to influence EPC number or function. Adapted from
Ref. [13].
with a more recent report of similarly diminished cells in women
with pre-eclampsia [34], but a definitive conclusion remains lack- Agent Effect on EPCs Mode of action
ing. Future investigations are needed to define the number and Angiotensin-II CAC senescence Angiotensin type 1 receptor
function of EPCs prospectively, prior to disease onset, and whether (AT1R)
Angiotensin-II Increases EPC numbers, C-kit-expression
identified distortions are a cause or consequence of this syndrome.
inhibitors migration
Erythropoietin Increases CAC number, NO-dependent
mobilization anti-
10. Clinical perspectives
apoptotic, NO-synthesis
Estradiol Mobilizes and encourages NO and MMP-9-dependent
Considering available evidence, it could be speculated that EPCs proliferation
are involved in the vascular wellbeing of the fetus, placenta and Nifedipine Increases CAC number, Unknown
perhaps even the expectant mother. From a maternal perspective, function, and resistance
to oxidative stress
EPCs may be required for systemic vascular adaptations to preg- Physical exercise Increases EPC number, NO-dependent, increases
nancy and uterine homeostasis. In pre-eclampsia it is possible that capacity inhibits apoptosis VEGF
deficiencies or functional impairments in EPCs, both CACs and PPAR-d agonists Stimulates CAC PI3K/Akt pathway.
ECFCs, may render a woman more susceptible to inflammatory or proliferation, mobilization.
Anti-apoptotic.
metabolic insults, thus exacerbating endothelial dysfunction. The
Prostaglandin E1 Enhances mobilization NO-dependent, Increased
fact that this dysfunction can persist beyond pregnancy, as defined and function CXCR4
epidemiologically, through an elevated risk of hypertension, coro- Statins Elevates EPCs in bone PI3K/Akt pathway
nary or cerebro-vascular disease [52], raises the idea that defective marrow and circulation.
EPCs are inherent and life-long. In this respect, an age-related Integrin-upregulation
Sildenafil Increases EPC numbers Unknown
decline in EPC numbers and efficiency has been noted, and
S66 I.P. Crocker, P.I. Sipos / Placenta 34, Supplement A, Trophoblast Research, Vol. 27 (2013) S62eS67

References [27] Sipos P, Davidge S, Baker P, Crocker I, Hubel C. The role of fetal endothelial
progenitor cells (EPCs) in placental vasculogenesis. Reproductive Sciences
2011;18(4):71A.
[1] Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation
[28] Verlohren S, Stepan H, Dechend R. Angiogenic growth factors in the diagnosis
of putative progenitor endothelial cells for angiogenesis. Science (New York,
and prediction of pre-eclampsia. Clinical Science (London) 2012;122(2):43e52.
NY) 1997;275(5302):964e7.
[29] Moyes AJ, Maldonado-Perez D, Gray GA, Denison FC. Enhanced angiogenic
[2] Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, et al. Evidence for circulating
capacity of human umbilical vein endothelial cells from women with pre-
bone marrow-derived endothelial cells. Blood 1998;92(2):362e7.
eclampsia. Reproductive Sciences (Thousand Oaks, California) 2011;18(4):
[3] Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, et al.
374e82.
Circulating endothelial progenitor cells, vascular function, and cardiovascular
[30] Gilabert R, Bellart J, Jove M, Miralles RM, Piera V. Endothelial cell lesion in
risk. The New England Journal of Medicine 2003;348(7):593e600.
preeclampsia. Morphofunctional study using umbilical endothelial cells. Gy-
[4] Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, et al. Circulating
necologic and Obstetric Investigation 1999;47(2):95e101.
endothelial progenitor cells and cardiovascular outcomes. The New England
[31] Wang Y, Gu Y, Granger DN, Roberts JM, Alexander JS. Endothelial junctional
Journal of Medicine 2005;353(10):999e1007.
protein redistribution and increased monolayer permeability in human um-
[5] Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, et al.
bilical vein endothelial cells isolated during preeclampsia. American Journal of
Human endothelial progenitor cells from type II diabetics exhibit impaired
Obstetrics and Gynecology 2002;186(2):214e20.
proliferation, adhesion, and incorporation into vascular structures. Circulation
[32] Remuzzi G, Marchesi D, Zoja C, Muratore D, Mecca G, Misiani R, et al. Reduced
2002;106(22):2781e6.
umbilical and placental vascular prostacyclin in severe pre-eclampsia. Pros-
[6] Grisar J, Aletaha D, Steiner CW, Kapral T, Steiner S, Seidinger D, et al. Depletion
taglandins 1980;20(1):105e10.
of endothelial progenitor cells in the peripheral blood of patients with
[33] Xia L, Zhou XP, Zhu JH, Xie XD, Zhang H, Wang XX, et al. Decrease and dys-
rheumatoid arthritis. Circulation 2005;111(2):204e11.
function of endothelial progenitor cells in umbilical cord blood with maternal
[7] Tsai TH, Chai HT, Sun CK, Yen CH, Leu S, Chen YL, et al. Obesity suppresses
pre-eclampsia. The Journal of Obstetrics and Gynaecology Research 2007;
circulating level and function of endothelial progenitor cells and heart func-
33(4):465e74.
tion. Journal of Translational Medicine 2012;10(1):137.
[34] Lin C, Rajakumar A, Plymire DA, Verma V, Markovic N, Hubel CA. Maternal
[8] Case J, Mead LE, Bessler WK, Prater D, White HA, Saadatzadeh MR, et al.
endothelial progenitor colony-forming units with macrophage characteristics
Human CD34þAC133þVEGFR-2þ cells are not endothelial progenitor cells
are reduced in preeclampsia. American Journal of Hypertension 2009;22(9):
but distinct, primitive hematopoietic progenitors. Experimental Hematology
1014e9.
2007;35(7):1109e18.
[35] Kwon JY, Maeng YS, Kwon YG, Kim YH, Kang MH, Park YW. Decreased
[9] Yoon CH, Hur J, Park KW, Kim JH, Lee CS, Oh IY, et al. Synergistic neo-
endothelial progenitor cells in umbilical cord blood in severe preeclampsia.
vascularization by mixed transplantation of early endothelial progenitor cells
Gynecologic and Obstetric Investigation 2007;64(2):103e8.
and late outgrowth endothelial cells: the role of angiogenic cytokines and
[36] Padavala S, Pope N, Baker P, Crocker I. An imbalance between vascular
matrix metalloproteinases. Circulation 2005;112(11):1618e27.
endothelial growth factor and its soluble receptor in placental villous explants
[10] Tilling L, Chowienczyk P, Clapp B. Progenitors in motion: mechanisms of
of intrauterine growth-restricted pregnancies. Journal of the Society for Gy-
mobilization of endothelial progenitor cells. British Journal of Clinical Phar-
necologic Investigation 2006;13(1):40e7.
macology 2009;68(4):484e92.
[37] Sipos P, Wareing M, Sibley C, Baker P, Crocker I. Potential role of fetal-derived
[11] Duda DG, Fukumura D, Jain RK. Role of eNOS in neovascularization: NO for
endothelial progenitor cells in placental vasculogenesis and IUGR. Repro-
endothelial progenitor cells. Trends in Molecular Medicine 2004;10(4):143e5.
ductive Sciences 2010;17(3):152A.
[12] Nakamura Y, Tajima F, Ishiga K, Yamazaki H, Oshimura M, Shiota G, et al.
[38] Hwang HS, Maeng YS, Park YW, Koos BJ, Kwon YG, Kim YH. Increased sen-
Soluble c-kit receptor mobilizes hematopoietic stem cells to peripheral blood
escence and reduced functional ability of fetal endothelial progenitor cells in
in mice. Experimental Hematology 2004;32(4):390e6.
pregnancies complicated by preeclampsia without intrauterine growth re-
[13] Sipos PI, Crocker IP, Hubel CA, Baker PN. Endothelial progenitor cells: their
striction. American Journal of Obstetrics and Gynecology 2008;199(3):259.
potential in the placental vasculature and related complications. Placenta
e1e7.
2009;31(1):1e10.
[39] Ligi I, Simoncini S, Tellier E, Vassallo PF, Sabatier F, Guillet B, et al. A switch
[14] Chan RW, Schwab KE, Gargett CE. Clonogenicity of human endometrial epi-
toward angiostatic gene expression impairs the angiogenic properties of
thelial and stromal cells. Biology of Reproduction 2004;70(6):1738e50.
endothelial progenitor cells in low birth weight preterm infants. Blood 2011;
[15] Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, et al. Bone
118(6):1699e709.
marrow origin of endothelial progenitor cells responsible for postnatal vas-
[40] Luppi P, Powers RW, Verma V, Edmunds L, Plymire D, Hubel CA. Maternal
culogenesis in physiological and pathological neovascularization. Circulation
circulating CD34þVEGFR-2þ and CD133þVEGFR-2þ progenitor cells increase
Research 1999;85(3):221e8.
during normal pregnancy but are reduced in women with preeclampsia.
[16] Masuda H, Kalka C, Takahashi T, Yoshida M, Wada M, Kobori M, et al. Estro-
Reproductive Sciences (Thousand Oaks, California) 2010;17(7):643e52.
gen-mediated endothelial progenitor cell biology and kinetics for physiolog-
[41] Buemi M, Allegra A, D’Anna R, Coppolino G, Crasci E, Giordano D, et al. Con-
ical postnatal vasculogenesis. Circulation Research 2007;101(6):598e606.
centration of circulating endothelial progenitor cells (EPC) in normal preg-
[17] Elsheikh E, Sylven C, Ericzon BG, Palmblad J, Mints M. Cyclic variability of stromal
nancy and in pregnant women with diabetes and hypertension. American
cell-derived factor-1 and endothelial progenitor cells during the menstrual cycle.
Journal of Obstetrics and Gynecology 2007;196(1):68.e1e6.
International Journal of Molecular Medicine 2011;27(2):221e6.
[42] Matsubara K, Abe E, Matsubara Y, Kameda K, Ito M. Circulating endothelial
[18] Robb AO, Mills NL, Smith IB, Short A, Tura-Ceide O, Barclay GR, et al. Influence
progenitor cells during normal pregnancy and pre-eclampsia. American
of menstrual cycle on circulating endothelial progenitor cells. Human
Journal of Reproductive Immunology 2006;56(2):79e85.
Reproduction (Oxford, England) 2009;24(3):619e25.
[43] Javed MJ, Mead LE, Prater D, Bessler WK, Foster D, Case J, et al. Endothelial colony
[19] Schaaps JP, Tsatsaris V, Goffin F, Brichant JF, Delbecque K, Tebache M, et al.
forming cells and mesenchymal stem cells are enriched at different gestational
Shunting the intervillous space: new concepts in human uteroplacental vas-
ages in human umbilical cord blood. Pediatric Research 2008;64(1):68e73.
cularization. American Journal of Obstetrics and Gynecology 2005;192(1):
[44] Sugawara J, Mitsui-Saito M, Hoshiai T, Hayashi C, Kimura Y, Okamura K. Cir-
323e32.
culating endothelial progenitor cells during human pregnancy. The Journal of
[20] Bulmer JN, Burton GJ, Collins S, Cotechini T, Crocker IP, Croy BA, et al. IFPA
Clinical Endocrinology and Metabolism 2005;90(3):1845e8.
meeting 2011 workshop report II: angiogenic signaling and regulation of fetal
[45] Gussin HA, Bischoff FZ, Hoffman R, Elias S. Endothelial precursor cells in the
endothelial function; placental and fetal circulation and growth; spiral artery
peripheral blood of pregnant women. Journal of the Society for Gynecologic
remodeling. Placenta 2011;33(Suppl.):S9e14.
Investigation 2002;9(6):357e61.
[21] Sipos P, Rens W, Schlecht H, Fen F, Wareing M, Baker P, et al. Fetal endothelial
[46] Iwakura A, Luedemann C, Shastry S, Hanley A, Kearney M, Aikawa R, et al.
progenitor cells transmigration to the maternal circulation and contribution
Estrogen-mediated, endothelial nitric oxide synthase-dependent mobilization
to the vasculature of the pregnant uterus. Reproductive Sciences 2012;19(3):
of bone marrow-derived endothelial progenitor cells contributes to reendo-
84A.
thelialization after arterial injury. Circulation 2003;108(25):3115e21.
[22] Ingram DA, Mead LE, Tanaka H, Meade V, Fenoglio A, Mortell K, et al. Iden-
[47] Hamada H, Kim MK, Iwakura A, Ii M, Thorne T, Qin G, et al. Estrogen receptors
tification of a novel hierarchy of endothelial progenitor cells using human
alpha and beta mediate contribution of bone marrow-derived endothelial
peripheral and umbilical cord blood. Blood 2004;104(9):2752e60.
progenitor cells to functional recovery after myocardial infarction. Circulation
[23] Maruyama T, Masuda H, Ono M, Kajitani T, Yoshimura Y. Human uterine stem/
2006;114(21):2261e70.
progenitor cells: their possible role in uterine physiology and pathology.
[48] Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D,
Reproduction (Cambridge, England) 2010;140(1):11e22.
Ramaswami P, et al. Aging, progenitor cell exhaustion, and atherosclerosis.
[24] Becker CM, Beaudry P, Funakoshi T, Benny O, Zaslavsky A, Zurakowski D, et al.
Circulation 2003;108(4):457e63.
Circulating endothelial progenitor cells are up-regulated in a mouse model of
[49] Menegazzo L, Albiero M, Avogaro A, Fadini GP. Endothelial progenitor cells in
endometriosis. The American Journal of Pathology 2011;178(4):1782e91.
diabetes mellitus. BioFactors (Oxford, England) 38(3):194e202.
[25] Masuda H, Matsuzaki Y, Hiratsu E, Ono M, Nagashima T, Kajitani T, et al. Stem
[50] de Groot K, Bahlmann FH, Sowa J, Koenig J, Menne J, Haller H, et al. Uremia
cell-like properties of the endometrial side population: implication in endo-
causes endothelial progenitor cell deficiency. Kidney International 2004;
metrial regeneration. PloS One 2010;5(4):e10387.
66(2):641e6.
[26] Gekas C, Rhodes KE, Van Handel B, Chhabra A, Ueno M, Mikkola HK. He-
[51] Sugawara J, Mitsui-Saito M, Hayashi C, Hoshiai T, Senoo M, Chisaka H, et al.
matopoietic stem cell development in the placenta. The International Journal
Decrease and senescence of endothelial progenitor cells in patients with
of Developmental Biology 2010;54(6e7):1089e98.
I.P. Crocker, P.I. Sipos / Placenta 34, Supplement A, Trophoblast Research, Vol. 27 (2013) S62eS67 S67

preeclampsia. The Journal of Clinical Endocrinology and Metabolism 2005; [53] Williamson K, Stringer S, Sipos P, Crocker I, Alexander M. Age-related mod-
90(9):5329e32. ification of heparan sulphate proteoglycans (HSPGs) on human endothelial
[52] Wilson BJ, Watson MS, Prescott GJ, Sunderland S, Campbell DM, Hannaford P, progenitor cells. Heart 2011;97(20):14e5.
et al. Hypertensive diseases of pregnancy and risk of hypertension and stroke [54] Wang F, Wang Y, Zhang L, Zou L. Gene modification with integrin-linked ki-
in later life: results from cohort study. British Medical Journal (Clinical nase improves function of endothelial progenitor cells in pre-eclampsia
Research Edition) 2003;326(7394):845. in vitro. Journal of Cellular Biochemistry 2011;112(11):3103e11.

Anda mungkin juga menyukai