Você está na página 1de 4

Placenta xxx (2016) 1e4

Contents lists available at ScienceDirect

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

Human trophoblast stem cells: Real or not real?


Ching-Wen Chang a, b, Mana M. Parast a, b, *
a
Department of Pathology, University of California San Diego, La Jolla, CA, USA
b
Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA

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

Article history: Abnormal trophoblast differentiation is the root cause of many placenta-based pregnancy complications,
Received 24 October 2016 including preeclampsia and fetal growth restriction. Human trophoblast differentiation is difcult to
Received in revised form study due to the lack of a stem cell model. Such a multipotent trophoblast stem (TS) cell, with the
30 December 2016
ability to differentiate into all trophoblast subtypes, has been derived from mouse blastocysts, but at-
Accepted 4 January 2017
tempts to derive similar human cells have failed. We consider here several possibilities for the TS cell
niche in the human placenta. Aside from discussion of such a niche in the pre-implantation blastocyst,
Keywords:
we discuss evidence for these TS cells residing in the post-implantation villous cytotrophoblast layer, or
Placenta
Blastocyst
even in the non-trophoblast portions, of the human placenta. It is our hope that recognition of the niche
Trophoblast stem cells would lead to successful derivation and in vitro establishment of such cells, which could then be
Trophoblast disseminated widely to the placental biology community for advancing the eld. Availability of self-
Progenitor cells renewing human TS cells, whose gene expression and environment could be manipulated, will pro-
vide a platform, not just for the study of pathophysiology of placental disease, but also for the discovery
of diagnostic biomarkers and therapeutic targets for common pregnancy complications.
2017 Elsevier Ltd. All rights reserved.

1. Introduction Knowledge of the TSC niche in the polar trophectoderm of the


mouse embryo led to their successful derivation almost 20 years
The concept of a stem cell niche was rst suggested by Schoeld ago [6]. However, isolation of similar cells from the human blas-
in 1978 to describe the bone-marrow microenvironment of he- tocyst has remained a challenge [7,8], due, at least in part, to the
matopoietic stem cells (HSCs) [1]. It was initially dened as the site lack of knowledge regarding the TSC niche during early human
of residence of stem cells in a particular anatomical tissue; how- development. Studies have pointed to both various gestational ages
ever, over the past 30 years, this denition has evolved and now and various compartments, where such human TSC might exist
includes a particular microenvironment which protects stem cells, [7e14]. In this review, we will discuss the various proposed niches
providing them with growth factors and extracellular matrix for human TSC (Fig. 1), highlighting the many questions that remain
components to maintain their stemness. Knowledge of the niche and the areas which require further study.
has led to derivation of various types of stem cells, including em-
bryonic stem cells (ESCs) and tissue-specic stem cells (such as
2. Trophectoderm of the preimplantation blastocyst
HSCs, and mesenchymal stem cells/MSCs), all of which have proven
useful as in vitro models for studying molecular mechanisms of
Trophoblast lineage specication begins with the formation of
lineage specication and organ development [2e4].
the blastocyst, where inner cell mass (ICM), the precursor to all
The human placenta has been dubbed the least understood
embryonic tissues as well as extraembryonic endoderm and
organ [5]; unfortunately, this statement also applies to the stem
mesoderm, separates from an outer layer of cells called tro-
cells which contribute to this organ, namely, trophoblast stem cells
phectoderm (TE), which are thought to give rise to all trophoblast
(TSCs) which give rise to the epithelial components of the placenta.
(extraembryonic ectoderm) subtypes after implantation [7]. TSC
were rst isolated from TE outgrowths of mouse blastocysts, using
a combination of broblast growth factor 4 (FGF4) and mouse
* Corresponding author. Sanford Consortium for Regenerative Medicine,
Department of Pathology, University of California San Diego, 9500 Gilman Drive,
embryonic broblast (MEFs) [6]; the latter secrete TGF-beta/activin
MC 0695, La Jolla, CA 92093, USA. as the required active components, and thus can be replaced by
E-mail address: mparast@ucsd.edu (M.M. Parast). media conditioned by the feeders [15]. Mouse TSCs are

http://dx.doi.org/10.1016/j.placenta.2017.01.003
0143-4004/ 2017 Elsevier Ltd. All rights reserved.

Please cite this article in press as: C.-W. Chang, M.M. Parast, Human trophoblast stem cells: Real or not real?, Placenta (2016), http://dx.doi.org/
10.1016/j.placenta.2017.01.003
2 C.-W. Chang, M.M. Parast / Placenta xxx (2016) 1e4

Fig. 1. Schematic diagram demonstrating potential trophoblast stem cell niches in the human placenta at various gestational stages. (A) In the preimplantation stage, embryonic
stem cells (hESC) lines have been derived from early blastocysts, which appear to show totipotency (the ability to differentiate into endoderm, mesoderm, ectoderm, and tro-
phectoderm, the latter dened as CDX2 cells). In the blastocyst-proper, two different cell lineages are observed: trophectoderm (TE) and inner cells mass (ICM). Unlike mouse TE, in
the human blastocyst, CDX2 expression initially overlaps with OCT4 in TE, where GATA3 is also expressed. (B) In the post-implantation placenta, potential trophoblast progenitor
cell populations include cytotrophoblast subpopulations, such as those which co-express p63, CDX2, and ELF5, as well as ITGA4 cells in the chorionic mesenchyme. Proximal cell
column trophoblast are proliferative, but deemed as committed precursors to the invasive extravillous trophoblast lineage. TBPC, trophoblast progenitor cell; CTB, cytotrophoblast;
STB, syncytiotrophoblast; EVT extravillous cytotrophoblast; MC, mesenchymal cell; FV, fetal vessel.

characterized by expression of transcription factors, CDX2, ELF5, embryos; these hESC lines appear to have a unique gene expression
and EOMES, all of which are required for maintenance of this and DNA methylation prole, indicating a higher competence to-
lineage in vivo [11,12]. In the absence of feeders and FGF4, the cells ward trophoblast differentiation [22]. However, due to the early
lose the above markers, and begin to differentiate [6]. Recent nature of the embryonic stage used for their derivation, they cannot
studies of human embryos, however, have shown that, of the above be widely distributed for study [22]. Attempts to derive TSC from
factors, only CDX2 is specically expressed in the pre-implantation human blastocyst-stage embryos, based on culture conditions used
TE [9,14]. In addition, unlike the mouse, human TE initially co- for mouse TSC derivation or variations thereof, have been unsuc-
expressed OCT4 along with CDX2, with OCT4 being conned to cessful [23]. A comparative study of FGF receptors has recently
the ICM only in later stage blastocysts (~6 days post fertilization) revealed that FGFR2, the main FGF receptor expressed in the mouse
[14]. More recently, Blakeley et al. have taken these ndings further blastocyst, is not expressed in the human blastocyst. Interestingly,
using single-cell RNAseq, showing that key mouse TE-associated and similar to ELF5, FGFR2 does appear to be expressed in the post-
genes, including Elf5, Eomes and Id2, are completely absent in hu- implantation cytotrophoblast [23]. Based on these latter observa-
man TE; conversely, genes highly expressed in human TE, including tions, it has been proposed that, more likely, the human TSC niche
CLDN10, TRIML1 and PLAC8, were absent in mouse TE [9]. Inter- may reside in the post-implantation placenta [23].
estingly, of these, CLDN10, a tight junction component, may
participate in TSC niche formation, as establishment of apicobasal 3. Postimplantation chorionic villi
cell polarity is a critical step in TE formation in mice [16,17]; also, a
related family member, CLDN11, is required for establishment of the Following implantation, placental villous development rapidly
spermatogonial stem cell niche in mice [18]. Further study is progresses from invagination of proliferating cytotrophoblast
needed to unravel functions of these genes in human TE, with (primary villi), to invasion of these structures by mesenchymal cells
respect to both trophoblast lineage specication and/or TSC (secondary villi), and subsequent formation of primitive fetal blood
maintenance. vessels within them (tertiary villi) by the fourth week of gestation
To further study early lineage commitment in the human em- [24,25]. At this stage, two distinct trophoblast subpopulations are
bryo, two groups recently recapitulated this process in vitro, observed: a proliferative mononuclear cytotrophoblast (CTB) shell
showing that early embryonic development can take place in the which is immediately adjacent to the villous mesenchyme, and a
absence of any maternal tissues [19,20]. They observed the blas- multinucleated syncytiotrophoblast (STB) which abuts the
tocysts to always attach on the side of the polar TE, the area where maternal vascular space (Fig. 1). Continuous proliferation of the CTB
trophoblast is nearest the inner cell mass; this is distinct from the shell at points of contact with the uterine decidua leads to forma-
same process in mouse, where mural TE initiate attachment [21]. At tion of a third trophoblast subpopulation, the invasive extravillous
this stage in the human embryo, TE was best dened by nuclear trophoblast (EVT) [24]. In these regions of anchoring columns,
expression of GATA3, although a variable amount of CDX2 expres- there is a progression of differentiation from a proliferative villous
sion was also present. Following attachment, the embryo attened CTB, to a proliferative proximal column EVT, to a non-proliferative
and the GATA3 TE acquired strong lamentous CK7 staining, fol- distal column EVT, nally into fully differentiated invasive EVT
lowed, after a few days, by appearance of multinucleated cells and (Fig. 1). The latter include at least two subtypes, based on their
induction of human chorionic gonadotropin-beta (hCGb) expres- localization: interstitial EVT which invade decidua singly or in
sion in the expanding TE. Taken together, these results indicate that groups, and endovascular EVT, which invade and remodel maternal
polar TE is critical during implantation and that trophoblast dif- spiral arterioles [24,25]. Along this differentiation pathway,
ferentiation, at least in these early stages, can be induced within the markers of CTB (p63, EGFR) are lost, and those of EVT (HLAG,
embryo itself, without input from maternal tissues [19,20]. MelCAM) are gained [24e26]. During this differentiation, an
Recently, derivation of embryonic stem cell (hESC) lines have elegant integrin switching is also observed, with loss of ITGA6, and
been reported from single blastomeres of 8-to-10-cell human gain of ITGA5 and ITGA1 [27e29].

Please cite this article in press as: C.-W. Chang, M.M. Parast, Human trophoblast stem cells: Real or not real?, Placenta (2016), http://dx.doi.org/
10.1016/j.placenta.2017.01.003
C.-W. Chang, M.M. Parast / Placenta xxx (2016) 1e4 3

The primary proposed TSC niche within these early post- for such cells. Of course, it is highly likely that other compartments
implantation villi is the CTB layer, where, aside from p63 [26], a house progenitor cells which can give rise to a more limited
subset of cells have been shown to co-express CDX2 [30] and ELF5 repertoire of trophoblast subtypes (i.e. CTB in second trimester
[13]. These cells have been proposed as TSC, based purely on through term gestation, continue to proliferate and contribute to
comparative marker expression in mouse TSC, and the rapid loss of the STB layer). It is also possible that later gestation placental tissue
this population by the end of the rst trimester, after which CTB retains a small but signicant subpopulation of true TSC, which,
differentiation into HLA-G EVT becomes more limited [31]. In under specic conditions of damage/stress, and along with signals
addition, villous CTB have also been shown to express FGFR2, the from the mesenchymal compartment, are able to expand and
same receptor expressed on mouse TSC, and even proliferate in regenerate functional placental tissue.
response to FGF4, in context of the oating villous explant model Much work remains to test the above hypotheses. While the
(i.e. with their underlying mesenchyme intact) [10]. However, pri- isolation of primary CTB from various gestation human placentae
mary isolated villous CTB, either as a whole or in part, have yet to be can be done relatively routinely, variable access to early gestation
successfully passaged in vitro continuously. tissues and our inability to maintain these cells in a proliferative
Many epithelial stem cell populations contain transiently state present the two most signicant limitations to the study of
amplifying progenitor cells, cells dened as committed pro- human trophoblast differentiation and placental development. A
genitors, able to expand the progenitor pool in tissues as needed more detailed evaluation of cell types within the human placenta,
[32]. In the human placenta, the villous CTB layer has been pro- both during early gestation and in various placental compartments
posed to contain such progenitor populations. In fact, as early as 8 (chorionic villi, chorion, and basal plate), is required in order to
weeks gestation, there is evidence for two populations of CTB understand the niche(s) for TSC and other trophoblast progenitor
progenitorsdone that gives rise to STB, and another that gives rise cells, before we are able to establish such cells in continuous cul-
to EVTddened by different survival charactertistics in the context ture. Finally, much work on the human placenta is done by isolated
of the villous explant [33]. It is possible that a bona de TSC, groups/laboratories; signicantly more coordination and collabo-
residing in early (5e8 week gestation) placenta, gives rise in late rative effort is required between groups which focus on human
rst trimester to these two distinct progenitor cell populations. A placental biology, to standardize nomenclature and phenotypic
recent study by Haider et al. [34] supports this latter hypothesis, denitions of trophoblast subpopulations, cell isolation procedures,
with the identication of a NOTCH1 villous CTB subpopulation as well as culture conditions, in order to truly advance this eld.
which give rise exclusively to EVT. While primary CTB remain the gold standard, human pluripo-
Another characteristic of stem cells, the ability to extrude the tent stem cells (hPSCs, referring to both embryonic stem cells/
uorescent dye Hoechst, has been used to isolate a side popula- hESCs and induced pluripotent stem cells/iPSCs) have also been
tion of cells from amongst villous CTB [35]. Based on their gene successfully used to study human trophoblast differentiation
expression prole, the side population cells were more closely [37,38]. Our own work has recently led to development of a two-
related to CTB than EVT, and their proportion did not change step protocol, which uses dened culture conditions to produce a
signicantly between 6 and 12 weeks of gestation [35]. Interest- uniform population of CDX/p63/EGFR CTB stem-like cells,
ingly, extracellular matrix (ECM)-related genes were signicantly which can be replated and further differentiated into STB- and EVT-
enriched within these side population cells [35], indicating that the like cells [30]. The advantage of these cells is the ability to model
ECM likely plays a crucial role for establishment and maintenance both normal and abnormal trophoblast differentiation, using iPSCs
of this potential stem cell niche. Additional studies are necessary to derived from individuals/placentae with diverse pregnancy out-
determine if these side population cells are truly multipotent TSC. comes. In addition, these cells could provide a platform for
screening potential drug therapies aimed at placenta-based preg-
4. Putative TSC in the non-trophoblast (mesenchymal) nancy disorders such as preeclampsia and fetal growth restriction.
portions of placenta At the same time, application of similar methodology for derivation
of iPSC has recently led to derivation of iTSCdconversion of
Finally, the non-trophoblast portion of the chorion has also been mouse broblasts to trophoblast stem cells using dened factors
proposed as a niche for TSC (Fig. 1) [8]. By using a combination of [39,40]. Although the latter technology has yet to be extended to
FGF and inhibitors of the activin/nodal pathway, Genbacev et al. derivation of similar human cells, there is much hope that such
were able to isolate and maintain a putative TSC from the chorionic advances in regenerative medicine can also be applied to placental
mesenchyme of rst and second trimester placentae [8]. This biology, providing ever more sophisticated in vitro models for the
nding is deemed controversial, as these cells are likely of extra- study of this crucial human organ, which in turn would serve as
embryonic mesoderm (ICM) origin, where all trophoblast are platforms for screening of potential therapeutics aimed at
thought to be derived from TE. Nevertheless, these ITGA4 cells improving placental function.
seemed to have the ability to differentiate into both STB and EVT,
although it was not clear whether they rst underwent a CTB Acknowledgements
precursor phase; in fact, in subsequent studies, they seem to pri-
marily differentiate into HLAG EVT [36]. In addition, based on their This work was supported by funds from a CIRM Physician-
gene expression prole, they appear to have characteristics of both Scientist Award (RN3-06396) and NIH/NICHD (R01-HD071100).
embryonic stem cells and trophoblast [8,36]. However, similar to The authors have no conict of interest to report.
TSC-like embryonic stem cells derived from single human blas-
tomeres [22], the properties of these cells remain to be indepen-
dently conrmed. References

[1] R. Schoeld, The relationship between the spleen colony-forming cell and the
5. Conclusions and perspectives haemopoietic stem cell, Blood Cells 4 (1e2) (1978) 7e25.
[2] A. Trounson, A rapidly evolving revolution in stem cell biology and medicine,
While multiple niches have been described for putative human Reprod. Biomed. Online 27 (6) (2013) 756e764.
[3] A. Bigas, C. Waskow, Blood stem cells: from beginning to end, Development
trophoblast stem/progenitor cells, the bulk of the data point to 143 (19) (2016) 3429e3433.
villous tissue in the early gestation placenta as the most likely niche [4] A. Keating, Mesenchymal stromal cells: new directions, Cell Stem Cell. 10 (6)

Please cite this article in press as: C.-W. Chang, M.M. Parast, Human trophoblast stem cells: Real or not real?, Placenta (2016), http://dx.doi.org/
10.1016/j.placenta.2017.01.003
4 C.-W. Chang, M.M. Parast / Placenta xxx (2016) 1e4

(2012) 709e716. [23] T. Kunath, Y. Yamanaka, J. Detmar, D. MacPhee, I. Caniggia, J. Rossant,


[5] A.E. Guttmacher, Y.T. Maddox, C.Y. Spong, The Human Placenta Project: A. Jurisicova, Developmental differences in the expression of FGF receptors
placental structure, development, and function in real time, Placenta 35 (5) between human and mouse embryos, Placenta 35 (12) (2014) 1079e1088.
(2014) 303e304. [24] J.L. James, A.M. Carter, L.W. Chamley, Human placentation from nidation to 5
[6] S. Tanaka, T. Kunath, A.K. Hadjantonakis, A. Nagy, J. Rossant, Promotion of weeks of gestation. Part I: what do we know about formative placental
trophoblast stem cell proliferation by FGF4, Science 282 (5396) (1998) development following implantation? Placenta 33 (5) (2012) 327e334.
2072e2075. [25] K. Benirschke, G.J. Burton, P. Kaufmann, Pathology of the Human Placenta,
[7] J. Rossant, Stem cells and lineage development in the mammalian blastocyst, sixth ed., Springer-Verlag, Berlin Heidelberg, 2012.
Reprod. Fertil. Dev. 19 (1) (2007) 111e118. [26] Y. Lee, K.R. Kim, F. McKeon, A. Yang, T.K. Boyd, C.P. Crum, M.M. Parast,
[8] O. Genbacev, M. Donne, M. Kapidzic, M. Gormley, J. Lamb, J. Gilmore, A unifying concept of trophoblastic differentiation and malignancy dened by
N. Larocque, G. Golden, T. Zdravkovic, M.T. McMaster, S.J. Fisher, Establish- biomarker expression, Hum. Pathol. 38 (7) (2007) 1003e1013.
ment of human trophoblast progenitor cell lines from the chorion, Stem Cells [27] C.H. Damsky, M.L. Fitzgerald, S.J. Fisher, Distribution patterns of extracellular
29 (9) (2011) 1427e1436. matrix components and adhesion receptors are intricately modulated during
[9] P. Blakeley, N.M. Fogarty, I. Del Valle, S.E. Wamaitha, T.X. Hu, K. Elder, P. Snell, rst trimester cytotrophoblast differentiation along the invasive pathway,
L. Christie, P. Robson, K.K. Niakan, Dening the three cell lineages of the hu- in vivo, J. Clin. Invest. 89 (1) (1992) 210e222.
man blastocyst by single-cell RNA-seq, Development 142 (18) (2015) [28] C.H. Damsky, C. Librach, K.H. Lim, M.L. Fitzgerald, M.T. McMaster,
3151e3165. M. Janatpour, Y. Zhou, S.K. Logan, S.J. Fisher, Integrin switching regulates
[10] D. Baczyk, C. Dunk, B. Huppertz, C. Maxwell, F. Reister, D. Giannoulias, normal trophoblast invasion, Development 120 (12) (1994) 3657e3666.
J.C. Kingdom, Bi-potential behaviour of cytotrophoblasts in rst trimester [29] Y. Zhou, C.H. Damsky, K. Chiu, J.M. Roberts, S.J. Fisher, Preeclampsia is asso-
chorionic villi, Placenta 27 (4e5) (2006) 367e374. ciated with abnormal expression of adhesion molecules by invasive cyto-
[11] R. John, M. Hemberger, A placenta for life, Reprod. Biomed. 25 (1) (2012) trophoblasts, J. Clin. Invest. 91 (3) (1993) 950e960.
5e11. Online. [30] M. Horii, Y. Li, A.K. Wakeland, D.P. Pizzo, K.K. Nelson, K. Sabatini, L.C. Laurent,
[12] R.M. Roberts, S.J. Fisher, Trophoblast stem cells, Biol. Reprod. 84 (3) (2011) Y. Liu, M.M. Parast, Human pluripotent stem cells as a model of trophoblast
412e421. differentiation in both normal development and disease, Proc. Natl. Acad. Sci.
[13] M. Hemberger, R. Udayashankar, P. Tesar, H. Moore, G.J. Burton, ELF5-enforced U. S. A. 113 (27) (2016) E3882eE3891.
transcriptional networks dene an epigenetically regulated trophoblast stem [31] M.T. McMaster, C.L. Librach, Y. Zhou, K.H. Lim, M.J. Janatpour, R. DeMars,
cell compartment in the human placenta, Hum. Mol. Genet. 19 (12) (2010) S. Kovats, C. Damsky, S.J. Fisher, Human placental HLA-G expression is
2456e2467. restricted to differentiated cytotrophoblasts, J. Immunol. 154 (8) (1995)
[14] K.K. Niakan, K. Eggan, Analysis of human embryos from zygote to blastocyst 3771e3778.
reveals distinct gene expression patterns relative to the mouse, Dev. Biol. 375 [32] C. Blanpain, V. Horsley, E. Fuchs, Epithelial stem cells: turning over new
(1) (2013) 54e64. leaves, Cell 128 (3) (2007) 445e458.
[15] A. Erlebacher, K.A. Price, L.H. Glimcher, Maintenance of mouse trophoblast [33] J.L. James, P.R. Stone, L.W. Chamley, Cytotrophoblast differentiation in the rst
stem cell proliferation by TGF-beta/activin, Dev. Biol. 275 (1) (2004) 158e169. trimester of pregnancy: evidence for separate progenitors of extravillous
[16] V.B. Alarcon, Cell polarity regulator PARD6B is essential for trophectoderm trophoblasts and syncytiotrophoblast, Reproduction 130 (1) (2005) 95e103.
formation in the preimplantation mouse embryo, Biol. Reprod. 83 (3) (2010) [34] S. Haider, G. Meinhardt, L. Saleh, C. Fiala, J. Pollheimer, M. Kno er, Notch1
347e358. controls development of the extravillous trophoblast lineage in the human
[17] Y. Marikawa, V.B. Alarcon, Establishment of trophectoderm and inner cell placenta, Proc. Natl. Acad. Sci. U. S. A. 113 (48) (2016) E7710eE7719.
mass lineages in the mouse embryo, Mol. Reprod. Dev. 76 (11) (2009) [35] J.L. James, D.G. Hurley, T.K. Gamage, T. Zhang, R. Vather, P. Pantham, P. Murthi,
1019e1032. L.W. Chamley, Isolation and characterisation of a novel trophoblast side-
[18] A. Gow, C.M. Southwood, J.S. Li, M. Pariali, G.P. Riordan, S.E. Brodie, J. Danias, population from rst trimester placentae, Reproduction 150 (5) (2015)
J.M. Bronstein, B. Kachar, R.A. Lazzarini, CNS myelin and sertoli cell tight 449e462.
junction strands are absent in Osp/claudin-11 null mice, Cell 99 (6) (1999) [36] O. Genbacev, N. Larocque, K. Ona, A. Prakobphol, T. Garrido-Gomez,
649e659. M. Kapidzic, A. Ba rcena, M. Gormley, S.J. Fisher, Integrin alpha4-positive hu-
[19] A. Deglincerti, G.F. Croft, L.N. Pietila, M. Zernicka-Goetz, E.D. Siggia, man trophoblast progenitors: functional characterization and transcriptional
A.H. Brivanlou, Self-organization of the in vitro attached human embryo, regulation, Hum. Reprod. 31 (6) (2016) 1300e1314.
Nature 533 (7602) (2016) 251e254. [37] T.G. Golos, M. Giakoumopoulos, B. Gerami-Naini, Review: trophoblast differ-
[20] M.N. Shahbazi, A. Jedrusik, S. Vuoristo, G. Recher, A. Hupalowska, V. Bolton, entiation from human embryonic stem cells, Placenta (34 Suppl) (2013)
N.M. Fogarty, A. Campbell, L.G. Devito, D. Ilic, Y. Khalaf, K.K. Niakan, S. Fishel, S56eS61.
M. Zernicka-Goetz, Self-organization of the human embryo in the absence of [38] Y. Li, M.M. Parast, BMP4 regulation of human trophoblast development, Int. J.
maternal tissues, Nat. Cell Biol. 18 (6) (2016) 700e708. Dev. Biol. 58 (2e4) (2014) 239e246.
[21] S. Lindenberg, P. Hyttel, A. Sjgren, T. Greve, A comparative study of attach- [39] H. Benchetrit, S. Herman, N. van Wietmarschen, T. Wu, K. Makedonski,
ment of human, bovine and mouse blastocysts to uterine epithelial mono- N. Maoz, N. Yom Tov, D. Stave, R. Lasry, V. Zayat, A. Xiao, P.M. Lansdorp,
layer, Hum. Reprod. 4 (4) (1989) 446e456. S. Sebban, Y. Buganim, Extensive nuclear reprogramming underlies lineage
[22] T. Zdravkovic, K.L. Nazor, N. Larocque, M. Gormley, M. Donne, N. Hunkapillar, conversion into functional trophoblast stem-like cells, Cell Stem Cell. 17 (5)
G. Giritharan, H.S. Bernstein, G. Wei, M. Hebrok, X. Zeng, O. Genbacev, (2015) 543e556.
A. Mattis, M.T. McMaster, A. Krtolica, D. Valbuena, C. Simo n, L.C. Laurent, [40] C. Kubaczka, C.E. Senner, M. Cierlitza, M.J. Arazo-Bravo, P. Kuckenberg,
J.F. Loring, S.J. Fisher, Human stem cells from single blastomeres reveal M. Peitz, M. Hemberger, H. Schorle, Direct induction of trophoblast stem cells
pathways of embryonic or trophoblast fate specication, Development 142 from murine broblasts, Cell Stem Cell. 17 (5) (2015) 557e568.
(23) (2015) 4010e4025.

Please cite this article in press as: C.-W. Chang, M.M. Parast, Human trophoblast stem cells: Real or not real?, Placenta (2016), http://dx.doi.org/
10.1016/j.placenta.2017.01.003

Você também pode gostar