@article{karakis_jabeen_britt_cordiner_mischler_li_miguel_rao_2023, title={Laminin switches terminal differentiation fate of human trophoblast stem cells under chemically defined culture conditions}, volume={299}, ISSN={["1083-351X"]}, DOI={10.1016/j.jbc.2023.104650}, abstractNote={Human trophoblast stem cells (hTSCs) have emerged as a powerful tool to model early placental development in vitro. Analogous to the epithelial cytotrophoblast in the placenta, hTSCs can differentiate into cells of the extravillous trophoblast (EVT) lineage or the multinucleate syncytiotrophoblast (STB). Here we present a chemically defined culture system for STB and EVT differentiation of hTSCs. Notably, in contrast to current approaches, we neither utilize forskolin for STB formation nor transforming growth factor-beta (TGFβ) inhibitors or a passage step for EVT differentiation. Strikingly, the presence of a single additional extracellular cue–laminin-111–switched the terminal differentiation of hTSCs from STB to the EVT lineage under these conditions. In the absence of laminin-111, STB formation occurred, with cell fusion comparable to that obtained with differentiation mediated by forskolin; however, in the presence of laminin-111, hTSCs differentiated to the EVT lineage. Protein expression of nuclear hypoxia-inducible factors (HIF1α and HIF2α) was upregulated during EVT differentiation mediated by laminin-111 exposure. A heterogeneous mixture of Notch1+ EVTs in colonies and HLA-G+ single-cell EVTs were obtained without a passage step, reminiscent of heterogeneity in vivo. Further analysis showed that inhibition of TGFβ signaling affected both STB and EVT differentiation mediated by laminin-111 exposure. TGFβ inhibition during EVT differentiation resulted in decreased HLA-G expression and increased Notch1 expression. On the other hand, TGFβ inhibition prevented STB formation. The chemically defined culture system for hTSC differentiation established herein facilitates quantitative analysis of heterogeneity that arises during hTSC differentiation and will enable mechanistic studies in vitro. Human trophoblast stem cells (hTSCs) have emerged as a powerful tool to model early placental development in vitro. Analogous to the epithelial cytotrophoblast in the placenta, hTSCs can differentiate into cells of the extravillous trophoblast (EVT) lineage or the multinucleate syncytiotrophoblast (STB). Here we present a chemically defined culture system for STB and EVT differentiation of hTSCs. Notably, in contrast to current approaches, we neither utilize forskolin for STB formation nor transforming growth factor-beta (TGFβ) inhibitors or a passage step for EVT differentiation. Strikingly, the presence of a single additional extracellular cue–laminin-111–switched the terminal differentiation of hTSCs from STB to the EVT lineage under these conditions. In the absence of laminin-111, STB formation occurred, with cell fusion comparable to that obtained with differentiation mediated by forskolin; however, in the presence of laminin-111, hTSCs differentiated to the EVT lineage. Protein expression of nuclear hypoxia-inducible factors (HIF1α and HIF2α) was upregulated during EVT differentiation mediated by laminin-111 exposure. A heterogeneous mixture of Notch1+ EVTs in colonies and HLA-G+ single-cell EVTs were obtained without a passage step, reminiscent of heterogeneity in vivo. Further analysis showed that inhibition of TGFβ signaling affected both STB and EVT differentiation mediated by laminin-111 exposure. TGFβ inhibition during EVT differentiation resulted in decreased HLA-G expression and increased Notch1 expression. On the other hand, TGFβ inhibition prevented STB formation. The chemically defined culture system for hTSC differentiation established herein facilitates quantitative analysis of heterogeneity that arises during hTSC differentiation and will enable mechanistic studies in vitro. The placenta is a complex fetal organ with a vast network of villi that ensures efficient exchange of nutrients and waste across the maternal-fetal interface. Epithelial cytotrophoblasts (CTBs) of the early human placenta give rise to all trophoblast cell types in the placenta (1Aplin J.D. Developmental cell biology of human villous trophoblast: current research problems.Int. J. Dev. Biol. 2010; 54: 323-329Crossref PubMed Scopus (95) Google Scholar, 2Knöfler M. Vasicek R. Schreiber M. Key regulatory transcription factors involved in placental trophoblast development - a review.Placenta. 2001; 22: S83-S92Crossref PubMed Scopus (45) Google Scholar, 3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar, 4Loregger T. Pollheimer J. Knöfler M. Regulatory transcription factors controlling function and differentiation of human trophoblast - a review.Placenta. 2003; 24: S104-S110Crossref PubMed Scopus (26) Google Scholar). CTBs undergo cell fusion to form the multinucleate syncytiotrophoblast (STB) that overlays the CTB layer of placental villi (1Aplin J.D. Developmental cell biology of human villous trophoblast: current research problems.Int. J. Dev. Biol. 2010; 54: 323-329Crossref PubMed Scopus (95) Google Scholar, 3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar). The STB is subsequently bathed in maternal blood at ∼10 weeks of gestation, when blood flow to the placenta is established (5Caniggia I. Winter J. Lye S.J. Post M. Oxygen and placental development during the first trimester: implications for the pathophysiology of pre-eclampsia.Placenta. 2000; 21: S25-S30Crossref PubMed Scopus (313) Google Scholar, 6Tuuli M.G. Longtine M.S. Nelson D.M. Review: oxygen and trophoblast biology - a source of controversy.Placenta. 2011; 32: S109-S118Crossref PubMed Scopus (120) Google Scholar, 7Zhou Y. Genbacev O. Damsky C.H. Fisher S.J. Oxygen regulates human cytotrophoblast differentiation and invasion: implications for endovascular invasion in normal pregnancy and in pre-eclampsia.J. Reprod. Immunol. 1998; 39: 197-213Crossref PubMed Scopus (94) Google Scholar). CTBs of placental villi anchored to the maternal decidua push through the syncytial layer and differentiate to extravillous trophoblasts (EVTs), first forming proliferative column trophoblasts adjacent to the villus tip (1Aplin J.D. Developmental cell biology of human villous trophoblast: current research problems.Int. J. Dev. Biol. 2010; 54: 323-329Crossref PubMed Scopus (95) Google Scholar, 3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar). At the distal end, column trophoblasts undergo an epithelial-to-mesenchymal transition (EMT) to become mature mesenchymal EVTs that invade the maternal decidua and parts of the myometrium (8Vićovac L. Aplin J.D. Epithelial-mesenchymal transition during trophoblast differentiation.Acta Anat. (Basel). 1996; 156: 202-216Crossref PubMed Scopus (209) Google Scholar, 9Thiery J.P. Epithelial-mesenchymal transitions in development and pathologies.Curr. Opin. Cell Biol. 2003; 15: 740-746Crossref PubMed Scopus (1456) Google Scholar, 10Dasilva-Arnold S. James J.L. Al-Khan A. Zamudio S. Illsley N.P. Differentiation of first trimester cytotrophoblast to extravillous trophoblast involves an epithelial-mesenchymal transition.Placenta. 2015; 36: 1412-1418Crossref PubMed Scopus (111) Google Scholar, 11Benirschke K. Burton G.J. Baergen R.N. Pathology of the Human Placenta.6th Ed. Springer, Berlin Heidelberg2012Crossref Scopus (64) Google Scholar). These invasive trophoblasts aid in remodeling maternal spiral arteries and play a critical role in establishing sufficient perfusion of the placenta with maternal blood (12Soares M.J. Iqbal K. Kozai K. Hypoxia and placental development.Birth Defects Res. 2017; 109: 1309-1329Crossref PubMed Scopus (89) Google Scholar, 13Harris L.K. Keogh R.J. Wareing M. Baker P.N. Cartwright J.E. Aplin J.D. et al.Invasive trophoblasts stimulate vascular smooth muscle cell apoptosis by a fas ligand-dependent mechanism.Am. J. Pathol. 2006; 169: 1863-1874Abstract Full Text Full Text PDF PubMed Scopus (122) Google Scholar, 14Lash G.E. Pitman H. Morgan H.L. Innes B.A. Agwu C.N. Bulmer J.N. Decidual macrophages: key regulators of vascular remodeling in human pregnancy.J. Leukoc. Biol. 2016; 100: 315-325Crossref PubMed Scopus (121) Google Scholar, 15Pijnenborg R. Vercruysse L. Hanssens M. The uterine spiral arteries in human pregnancy: facts and controversies.Placenta. 2006; 27: 939-958Crossref PubMed Scopus (873) Google Scholar). Many pregnancy complications including preeclampsia, fetal growth restriction, miscarriage, and stillbirth are often a result of impaired arterial remodeling and evidence points to improper EVT differentiation and invasion as a major contributor to these pathologies (3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar, 16Raymond D. Peterson E. A critical review of early-onset and late-onset preeclampsia.Obstet. Gynecol. Surv. 2011; 66: 497-506Crossref PubMed Scopus (376) Google Scholar, 17Prossler J. Chen Q. Chamley L. James J.L. The relationship between TGFβ, low oxygen and the outgrowth of extravillous trophoblasts from anchoring villi during the first trimester of pregnancy.Cytokine. 2014; 68: 9-15Crossref PubMed Scopus (34) Google Scholar, 18Arnholdt H. Meisel F. Fandrey K. Löhrs U. Proliferation of villous trophoblast of the human placenta in normal and abnormal pregnancies.Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 1991; 60: 365-372Crossref PubMed Scopus (148) Google Scholar, 19Silver R.M. Examining the link between placental pathology, growth restriction, and stillbirth.Best Pract. Res. Clin. Obstet. Gynaecol. 2018; 49: 89-102Crossref PubMed Scopus (35) Google Scholar, 20Romero R. Kusanovic J.P. Chaiworapongsa T. Hassan S.S. Placental bed disorders in preterm labor, preterm PROM, spontaneous abortion and abruptio placentae.Best Pract. Res. Clin. Obstet. Gynaecol. 2011; 25: 313-327Crossref PubMed Scopus (144) Google Scholar). For instance, quantitative analyses of images from preeclamptic placenta biopsies demonstrate shallow trophoblast invasion compared with healthy placenta (21Naicker T. Khedun S.M. Moodley J. Pijnenborg R. Quantitative analysis of trophoblast invasion in preeclampsia.Acta Obstet. Gynecol. Scand. 2003; 82: 722-729Crossref PubMed Scopus (153) Google Scholar). Previous studies have also suggested that increased apoptosis and an inability of CTBs to differentiate toward the EVT lineage may underlie shallow trophoblast invasion in preeclampsia (22Zhou Y. Damsky C.H. Fisher S.J. Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype: one cause of defective endovascular invasion in this syndrome?.J. Clin. Invest. 1997; 99: 2152-2164Crossref PubMed Scopus (815) Google Scholar, 23Zhou Y. Gormley M.J. Hunkapiller N.M. Kapidzic M. Stolyarov Y. Feng V. et al.Reversal of gene dysregulation in cultured cytotrophoblasts reveals possible causes of preeclampsia.J. Clin. Invest. 2013; 123: 2862-2872Crossref PubMed Scopus (97) Google Scholar). Yet, despite the importance of EVT differentiation in placental health and pathology, molecular mechanisms underlying EVT differentiation and maturation to mesenchymal invasive EVTs remain poorly understood. Mechanistic studies on early human placental development have been impeded primarily due to lack of suitable model systems. There are substantial restrictions on research with fetal tissue and human embryos and significant differences between placental development in common experimental animals and humans (24Boer G.J. Ethical guidelines for the use of human embryonic or fetal tissue for experimental and clinical neurotransplantation and research.J. Neurol. 1994; 242: 1-13Crossref PubMed Scopus (106) Google Scholar, 25Pera M.F. Human embryo research and the 14-day rule.Development. 2017; 144: 1923-1925Crossref PubMed Scopus (52) Google Scholar, 26Soncin F. Khater M. To C. Pizzo D. Farah O. Wakeland A. et al.Comparative analysis of mouse and human placentae across gestation reveals species-specific regulators of placental development.Development. 2018; 145dev156273Google Scholar, 27Lee K.Y. Demayo F.J. Animal models of implantation.Reproduction. 2004; 128: 679-695Crossref PubMed Scopus (213) Google Scholar, 28Malassine A. Frendo J.-L. Evain-Brion D. A comparison of placental development and endocrine functions between the human and mouse model.Hum. Reprod. Update. 2003; 9: 531-539Crossref PubMed Scopus (350) Google Scholar, 29Kunath T. Yamanaka Y. Detmar J. MacPhee D. Caniggia I. Rossant J. et al.Developmental differences in the expression of FGF receptors between human and mouse embryos.Placenta. 2014; 35: 1079-1088Crossref PubMed Scopus (73) Google Scholar). Additionally, there are significant differences in the transcriptome profiles of immortalized cell lines and primary trophoblasts (30Bilban M. Tauber S. Haslinger P. Pollheimer J. Saleh L. Pehamberger H. et al.Trophoblast invasion: assessment of cellular models using gene expression signatures.Placenta. 2010; 31: 989-996Crossref PubMed Scopus (153) Google Scholar). Further, immortalized cell lines do not model the heterogeneity of cell types observed during trophoblast differentiation in vivo (31Hannan N.J. Paiva P. Dimitriadis E. Salamonsen L.A. Models for study of human embryo implantation: choice of cell lines?.Biol. Reprod. 2010; 82: 235-245Crossref PubMed Scopus (237) Google Scholar). In this context, human trophoblast stem cells (hTSCs) derived from early gestation primary placental samples and blastocysts have emerged as powerful in vitro models of early human placental development (32Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. et al.Derivation of human trophoblast stem cells.Cell Stem Cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (432) Google Scholar). hTSCs, which model the CTB in vivo, can be maintained in cell culture and differentiate into STB or EVTs. In recent work, others and we have also shown that hTSCs can be derived from human pluripotent stem cells, including induced pluripotent stem cells (33Yu L. Wei Y. Duan J. Schmitz D.A. Sakurai M. Wang L. et al.Blastocyst-like structures generated from human pluripotent stem cells.Nature. 2021; 591: 620-626Crossref PubMed Scopus (190) Google Scholar, 34Amita M. Adachi K. Alexenko A.P. Sinha S. Schust D.J. Schulz L.C. et al.Complete and unidirectional conversion of human embryonic stem cells to trophoblast by BMP4.Proc. Natl. Acad. Sci. U. S. A. 2013; 110: E1212-E1221Crossref PubMed Scopus (184) Google Scholar, 35Castel G. Meistermann D. Bretin B. Firmin J. Blin J. Loubersac S. et al.Induction of human trophoblast stem cells from somatic cells and pluripotent stem cells.Cell Rep. 2020; 33: 108419Abstract Full Text Full Text PDF PubMed Scopus (84) Google Scholar, 36Wei Y. Wang T. Ma L. Zhang Y. Zhao Y. Lye K. et al.Efficient derivation of human trophoblast stem cells from primed pluripotent stem cells.Sci. Adv. 2021; 7eabf4416Crossref Scopus (37) Google Scholar, 37Dong C. Beltcheva M. Gontarz P. Zhang B. Popli P. Fischer L.A. et al.Derivation of trophoblast stem cells from naïve human pluripotent stem cells.Elife. 2020; 9e52504Crossref Scopus (143) Google Scholar, 38Mischler A. Karakis V. Mahinthakumar J. Carberry C.K. Miguel A.S. Rager J.E. et al.Two distinct trophectoderm lineage stem cells from human pluripotent stem cells.J. Biol. Chem. 2021; 296: 100386Abstract Full Text Full Text PDF PubMed Scopus (29) Google Scholar, 39Tiruthani K. Sarkar P. Rao B. Trophoblast differentiation of human embryonic stem cells.Biotechnol. J. 2013; 8: 421-433Crossref PubMed Scopus (9) Google Scholar, 40Sarkar P. Randall S.M. Collier T.S. Nero A. Russell T.A. Muddiman D.C. et al.Activin/nodal signaling switches the terminal fate of human embryonic stem cell-derived trophoblasts.J. Biol. Chem. 2015; 290: 8834-8848Abstract Full Text Full Text PDF PubMed Scopus (18) Google Scholar, 41Cinkornpumin J.K. Kwon S.Y. Guo Y. Hossain I. Sirois J. Russett C.S. et al.Naive human embryonic stem cells can give rise to cells with a trophoblast-like transcriptome and Methylome.Stem Cell Rep. 2020; 15: 198-213Abstract Full Text Full Text PDF PubMed Scopus (88) Google Scholar) and villous cytotrophoblasts from term placentas (42Wang L.J. Chen C.P. Lee Y.S. Ng P.S. Chang G.D. Pao Y.H. et al.Functional antagonism between ΔNp63α and GCM1 regulates human trophoblast stemness and differentiation.Nat. Commun. 2022; 13: 1-16PubMed Google Scholar), raising the exciting prospect of investigating pathological trophoblast development using somatic tissues (e.g., placenta) obtained at birth (42Wang L.J. Chen C.P. Lee Y.S. Ng P.S. Chang G.D. Pao Y.H. et al.Functional antagonism between ΔNp63α and GCM1 regulates human trophoblast stemness and differentiation.Nat. Commun. 2022; 13: 1-16PubMed Google Scholar, 43Sheridan M.A. Yang Y. Jain A. Lyons A.S. Yang P. Brahmasani S.R. et al.Early onset preeclampsia in a model for human placental trophoblast.Proc. Natl. Acad. Sci. U. S. A. 2019; 116: 4336-4345Crossref PubMed Scopus (42) Google Scholar). Nevertheless, current protocols for hTSC differentiation to EVT and STB limit mechanistic studies. Most current protocols for in vitro differentiation of hTSCs include transforming growth factor-beta (TGFβ) inhibitors during EVT differentiation (32Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. et al.Derivation of human trophoblast stem cells.Cell Stem Cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (432) Google Scholar, 44Io S. Iemura Y. Takashima Y. Optimized protocol for naive human pluripotent stem cell-derived trophoblast induction.STAR Protoc. 2021; 2: 100921Crossref PubMed Scopus (2) Google Scholar, 45Perez-Garcia V. Lea G. Lopez-Jimenez P. Okkenhaug H. Burton G.J. Moffett A. et al.BAP1/ASXL complex modulation regulates epithelial-mesenchymal transition during trophoblast differentiation and invasion.Elife. 2021; 10e63254Crossref PubMed Scopus (14) Google Scholar, 46Bai T. Peng C.Y. Aneas I. Sakabe N. Requena D.F. Billstrand C. et al.Establishment of human induced trophoblast stem-like cells from term villous cytotrophoblasts.Stem Cell Res. 2021; 56: 102507Crossref PubMed Scopus (11) Google Scholar, 47Karvas R.M. Khan S.A. Verma S. Yin Y. Kulkarni D. Dong C. et al.Stem-cell-derived trophoblast organoids model human placental development and susceptibility to emerging pathogens.Cell Stem Cell. 2022; 29: 810-825.e8Abstract Full Text Full Text PDF PubMed Scopus (32) Google Scholar). However, this is problematic because TGFβ plays an important role in EVT differentiation and dysregulation of TGFβ signaling can result in placental pathologies (11Benirschke K. Burton G.J. Baergen R.N. Pathology of the Human Placenta.6th Ed. Springer, Berlin Heidelberg2012Crossref Scopus (64) Google Scholar, 17Prossler J. Chen Q. Chamley L. James J.L. The relationship between TGFβ, low oxygen and the outgrowth of extravillous trophoblasts from anchoring villi during the first trimester of pregnancy.Cytokine. 2014; 68: 9-15Crossref PubMed Scopus (34) Google Scholar, 20Romero R. Kusanovic J.P. Chaiworapongsa T. Hassan S.S. Placental bed disorders in preterm labor, preterm PROM, spontaneous abortion and abruptio placentae.Best Pract. Res. Clin. Obstet. Gynaecol. 2011; 25: 313-327Crossref PubMed Scopus (144) Google Scholar, 48Lala P.K. Graham C.H. Mechanisms of trophoblast invasiveness and their control: the role of proteases and protease inhibitors.Cancer Metastasis Rev. 1990; 9: 369-379Crossref PubMed Scopus (241) Google Scholar, 49Cheng J.C. Chang H.M. Leung P.C.K. Transforming growth factor-β1 inhibits trophoblast cell invasion by inducing snail-mediated down-regulation of vascular endothelial-cadherin protein.J. Biol. Chem. 2013; 288: 33181-33192Abstract Full Text Full Text PDF PubMed Scopus (87) Google Scholar, 50Pollheimer J. Knöfler M. Signalling pathways regulating the invasive differentiation of human trophoblasts: a review.Placenta. 2005; 26: S21-S30Crossref PubMed Scopus (169) Google Scholar, 51Xu J. Sivasubramaniyam T. Yinon Y. Tagliaferro A. Ray J. Nevo O. et al.Aberrant TGFβ signaling contributes to altered trophoblast differentiation in preeclampsia.Endocrinology. 2016; 157: 883-899Crossref PubMed Scopus (35) Google Scholar, 52Graham C.H. Lala P.K. Mechanism of control of trophoblast invasion in situ.J. Cell Physiol. 1991; 148: 228-234Crossref PubMed Scopus (321) Google Scholar, 53Haider S. Kunihs V. Fiala C. Pollheimer J. Knöfler M. Expression pattern and phosphorylation status of Smad2/3 in different subtypes of human first trimester trophoblast.Placenta. 2017; 57: 17-25Crossref PubMed Scopus (17) Google Scholar, 54Haider S. Lackner A.I. Dietrich B. Kunihs V. Haslinger P. Meinhardt G. et al.Transforming growth factor-β signaling governs the differentiation program of extravillous trophoblasts in the developing human placenta.Proc. Natl. Acad. Sci. U. S. A. 2022; 119e2120667119Crossref PubMed Scopus (13) Google Scholar). Inhibition of TGFβ during EVT differentiation precludes investigation into the role of TGFβ signaling during normal or pathological trophoblast differentiation. Similarly, STB differentiation in vitro predominantly relies on the use of forskolin to induce cell fusion. Forskolin raises cyclic AMP concentrations causing upregulation of GCM1, which controls expression of fusion genes, syncytin-1 and syncytin-2 (55Chang C.-W. Chang G.-D. Chen H. A novel cyclic AMP/Epac1/CaMKI signaling cascade promotes GCM1 desumoylation and placental cell fusion.Mol. Cell. Biol. 2011; 31: 3820-3831Crossref PubMed Scopus (39) Google Scholar). The use of forskolin impedes studies on extracellular cues regulating STB differentiation. Finally, it is important to note that heterogeneity of cell types is a central feature of EVT differentiation in vivo. Initially, epithelial CTBs form proliferative proximal cell columns that express column markers MYC, VE-cadherin, Notch1, as well as CTB markers EGFR and ITGA6 (3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar, 8Vićovac L. Aplin J.D. Epithelial-mesenchymal transition during trophoblast differentiation.Acta Anat. (Basel). 1996; 156: 202-216Crossref PubMed Scopus (209) Google Scholar, 11Benirschke K. Burton G.J. Baergen R.N. Pathology of the Human Placenta.6th Ed. Springer, Berlin Heidelberg2012Crossref Scopus (64) Google Scholar, 32Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. et al.Derivation of human trophoblast stem cells.Cell Stem Cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (432) Google Scholar, 50Pollheimer J. Knöfler M. Signalling pathways regulating the invasive differentiation of human trophoblasts: a review.Placenta. 2005; 26: S21-S30Crossref PubMed Scopus (169) Google Scholar, 56Rosati E. Sabatini R. Rampino G. Tabilio A. Di Ianni M. Fettucciari K. et al.Constitutively activated notch signaling is involved in survival and apoptosis resistance of B-CLL cells.Blood. 2009; 113: 856-865Crossref PubMed Scopus (250) Google Scholar, 57Haider S. Meinhardt G. Saleh L. Kunihs V. Gamperl M. Kaindl U. et al.Self-renewing trophoblast organoids recapitulate the developmental program of the early human placenta.Stem Cell Rep. 2018; 11: 537-551Abstract Full Text Full Text PDF PubMed Scopus (207) Google Scholar, 58Haider S. Meinhardt G. Velicky P. Otti G.R. Whitley G. Fiala C. et al.Notch signaling plays a critical role in motility and differentiation of human first-trimester cytotrophoblasts.Endocrinology. 2014; 155: 263-274Crossref PubMed Scopus (46) Google Scholar, 59Haider S. Meinhardt G. Saleh L. Fiala C. Pollheimer J. Knöfler M. Notch1 controls development of the extravillous trophoblast lineage in the human placenta.Proc. Natl. Acad. Sci. U. S. A. 2016; 113: E7710-E7719Crossref PubMed Scopus (88) Google Scholar, 60Meinhardt G. Haider S. Haslinger P. Proestling K. Fiala C. Pollheimer J. et al.Wnt-dependent T-cell factor-4 controls human etravillous trophoblast motility.Endocrinology. 2014; 155: 1908-1920Crossref PubMed Scopus (46) Google Scholar, 61Hunkapiller N.M. Gasperowicz M. Kapidzic M. Plaks V. Maltepe E. Kitajewski J. et al.A role for notch signaling in trophoblast endovascular invasion and in the pathogenesis of pre-eclampsia.Development. 2011; 138: 2987-2998Crossref PubMed Scopus (130) Google Scholar, 62Plessl K. Haider S. Fiala C. Pollheimer J. Knöfler M. Expression pattern and function of notch2 in different subtypes of first trimester cytotrophoblast.Placenta. 2015; 36: 365-371Crossref PubMed Scopus (17) Google Scholar, 63Meinhardt G. Kaltenberger S. Fiala C. Knöfler M. Pollheimer J. ERBB2 gene amplification increases during the transition of proximal EGFR+ to distal HLA-G+ first trimester cell column trophoblasts.Placenta. 2015; 36: 803-808Crossref PubMed Scopus (16) Google Scholar, 64Pawar S.C. Demetriou M.C. Nagle R.B. Bowden G.T. Cress A.E. Integrin α6 cleavage: a novel modification to modulate cell migration.Exp. Cell Res. 2007; 313: 1080-1089Crossref PubMed Scopus (49) Google Scholar). Differentiating EVTs at the distal end of the epithelial column remain proliferative; however, they begin to express ITGA5 and low levels of the mature EVT marker HLA-G and lose CTB marker expression (11Benirschke K. Burton G.J. Baergen R.N. Pathology of the Human Placenta.6th Ed. Springer, Berlin Heidelberg2012Crossref Scopus (64) Google Scholar, 57Haider S. Meinhardt G. Saleh L. Kunihs V. Gamperl M. Kaindl U. et al.Self-renewing trophoblast organoids recapitulate the developmental program of the early human placenta.Stem Cell Rep. 2018; 11: 537-551Abstract Full Text Full Text PDF PubMed Scopus (207) Google Scholar, 62Plessl K. Haider S. Fiala C. Pollheimer J. Knöfler M. Expression pattern and function of notch2 in different subtypes of first trimester cytotrophoblast.Placenta. 2015; 36: 365-371Crossref PubMed Scopus (17) Google Scholar, 65Mccormick J. Whitley G.S.J. Le Bouteiller P. Cartwright J.E. Soluble HLA-G regulates motility and invasion of the trophoblast-derived cell line SGHPL-4.Hum. Reprod. 2009; 24: 1339-1345Crossref PubMed Scopus (35) Google Scholar). Subsequently, when these distal EVTs undergo EMT, they gain expression of ITGA1, express higher levels of HLA-G, and lose expression of markers that characterize the column trophoblasts (3Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: key molecular mechanisms and model systems.Cell. Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (311) Google Scholar, 66Fukushima K. Miyamoto S. Tsukimori K. Kobayashi H. Seki H. Takeda S. et al.Tumor necrosis factor and vascular endothelial growth factor induce endothelial integrin repertories, regulating endovascular differentiation and apoptosis in a human extravillous trophoblast cell Line1.Biol. Reprod. 2005; 73: 172-179Crossref PubMed Scopus (52) Google Scholar, 67Natenzon A. McFadden P. DaSilva-Arnold S.C. Zamudio S. Illsley N.P. Diminished trophoblast differentiation in early onset preeclampsia.Placenta. 2022; 120: 25-31Crossref PubMed Scopus (4) Google Scholar). Current protocols for EVT differentiation that include a passage step (32Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. et al.Derivation of human trophoblast stem cells.Cell Stem Cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (432) Google Scholar, 47Karvas R.M. Khan S.A. Verma S. Yin Y. Kulkarni D. Dong C. et al.Stem-cell-derived trophoblast organoids model human placental development and susceptibility to emerging pathogens.Cell Stem Cell. 2022; 29: 810-825.e8Abstract Full Text Full Text PDF PubMed Scopus (32) Google Scholar) do not capture EVT heterogeneity and the sequential nature of CTB differentiation as mature mesenchymal EVTs are formed. Here we present chemically defined culture conditions for differentiation of placenta- and hiPSC-derived hTSCs to EVTs and STB. Notably, our conditions do not involve a passage step and exclude forskolin and TGFβ inhibition during STB and EVT differentiation, respectively. Under these culture conditions, we identified laminin-111-mediated upregulation of hypoxia-inducible factor-alpha (HIFα) as the critical input that switches differentiation hTSCs from STB to the EVT lineage. We also investigated the effect of inhibiting TGFβ signaling on EVT and STB differentiation. Placenta-derived CT29 and CT30 hTSCs and hiPSC-derived SC102A-1 hTSCs were cultured in trophoblast stem cell medium (TSCM) as described previously (32Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. et al.Derivation of human trophoblast stem cells.Cell Stem Cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (432) Google Scholar, 38Mischler A. Karakis V. Mahinthakumar J. Carberry C.K. Miguel A.S. Rager J.E. et al.Two distinct trophectoderm lineage stem cells from human pluripotent stem cells.J. Biol. Chem. 2021; 296: 100386Abstract Full Text Full Text PDF PubMed Scopus (29) Google Scholar). Differentiation was induced by passaging hTSCs into a defined trophoblast differentiation medium (DTDM) supplemented with epidermal growth factor (EGF) and the ROCK inhibitor, Y-27632, at passage for 2 days, and culturing them for an additional 4 days in DTDM (Fig. 1A). Upon passage, we initially observed an increase in cell number, but b}, number={5}, journal={JOURNAL OF BIOLOGICAL CHEMISTRY}, author={Karakis, Victoria and Jabeen, Mahe and Britt, John W. and Cordiner, Abigail and Mischler, Adam and Li, Feng and Miguel, Adriana San and Rao, Balaji M.}, year={2023}, month={May} } @article{mischler_karakis_mahinthakumar_carberry_san miguel_rager_fry_rao_2021, title={Two distinct trophectoderm lineage stem cells from human pluripotent stem cells}, volume={296}, ISSN={["1083-351X"]}, url={http://dx.doi.org/10.1016/j.jbc.2021.100386}, DOI={10.1016/j.jbc.2021.100386}, abstractNote={The trophectoderm layer of the blastocyst-stage embryo is the precursor for all trophoblast cells in the placenta. Human trophoblast stem (TS) cells have emerged as an attractive tool for studies on early trophoblast development. However, the use of TS cell models is constrained by the limited genetic diversity of existing TS cell lines and restrictions on using human fetal tissue or embryos needed to generate additional lines. Here we report the derivation of two distinct stem cell types of the trophectoderm lineage from human pluripotent stem cells. Analogous to villous cytotrophoblasts in vivo, the first is a CDX2- stem cell comparable with placenta-derived TS cells—they both exhibit identical expression of key markers, are maintained in culture and differentiate under similar conditions, and share high transcriptome similarity. The second is a CDX2+ stem cell with distinct cell culture requirements, and differences in gene expression and differentiation, relative to CDX2- stem cells. Derivation of TS cells from pluripotent stem cells will significantly enable construction of in vitro models for normal and pathological placental development. The trophectoderm layer of the blastocyst-stage embryo is the precursor for all trophoblast cells in the placenta. Human trophoblast stem (TS) cells have emerged as an attractive tool for studies on early trophoblast development. However, the use of TS cell models is constrained by the limited genetic diversity of existing TS cell lines and restrictions on using human fetal tissue or embryos needed to generate additional lines. Here we report the derivation of two distinct stem cell types of the trophectoderm lineage from human pluripotent stem cells. Analogous to villous cytotrophoblasts in vivo, the first is a CDX2- stem cell comparable with placenta-derived TS cells—they both exhibit identical expression of key markers, are maintained in culture and differentiate under similar conditions, and share high transcriptome similarity. The second is a CDX2+ stem cell with distinct cell culture requirements, and differences in gene expression and differentiation, relative to CDX2- stem cells. Derivation of TS cells from pluripotent stem cells will significantly enable construction of in vitro models for normal and pathological placental development. Specification of the trophectoderm and the inner cell mass is the first differentiation event during human embryonic development. The trophectoderm mediates blastocyst implantation in the uterus and is the precursor to all trophoblast cells in the placenta. Upon embryo implantation, the trophectoderm forms the cytotrophoblast (CTB), a putative stem cell that can differentiate to form the two major cell types in the placenta, the extravillous trophoblast (EVT) and the syncytiotrophoblast (STB) (1Bischof P. Irminger-Finger I. The human cytotrophoblastic cell, a mononuclear chameleon.Int. J. Biochem. Cel. Biol. 2005; 37: 1-16Crossref PubMed Scopus (125) Google Scholar, 2Benirschke Kurt. Baergen R.N. Burton G. Graham J. Pathology of the Human Placenta [electronic Resource]. Springer, Heidelberg2012Crossref Scopus (48) Google Scholar). The EVTs are involved in remodeling of uterine arteries, which is critical to ensure adequate perfusion of the placenta with maternal blood, whereas the multinucleated STB mediates the nutrient and gas exchange at the maternal–fetal interface (3Yabe S. Alexenko A.P. Amita M. Yang Y. Schust D.J. Sadovsky Y. Ezashi T. Roberts R.M. Comparison of syncytiotrophoblast generated from human embryonic stem cells and from term placentas.Proc. Natl. Acad. Sci. U. S. A. 2016; 113: E2598-E2607Crossref PubMed Scopus (68) Google Scholar, 4Moser G. Orendi K. Gauster M. Siwetz M. Helige C. Huppertz B. The art of identification of extravillous trophoblast.Placenta. 2011; 32: 197-199Crossref PubMed Scopus (29) Google Scholar). Abnormalities in trophoblast development are associated with pregnancy-related pathologies such as miscarriage, preeclampsia, and placenta accreta. Yet, despite its relevance to maternal and fetal health, constraints on research with human embryos and early fetal tissue impede mechanistic insight into early trophoblast development. Trophoblast stem (TS) cells derived from first-trimester human placental samples and blastocyst-stage embryos have emerged as an attractive in vitro model system for early human trophoblast (5Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. Kabayama Y. Suyama M. Sasaki H. Arima T. Derivation of human trophoblast stem cells.Cell stem cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (233) Google Scholar). However, restricted accessibility of embryos and placental samples from early gestation and low genetic diversity of existing cell lines limit the use of this model. In contrast, human pluripotent stem cells (hPSCs) are a more accessible source for generating in vitro models of human trophoblast. Of more importance, unlike early gestation primary samples where the projected pregnancy outcome is uncertain, human induced pluripotent stem cells (hiPSCs) can potentially provide models of validated normal and pathological trophoblast development (6Sheridan M.A. Yang Y. Jain A. Lyons A.S. Yang P. Brahmasani S.R. Dai A. Tian Y. Ellersieck M.R. Tuteja G. Schust D.J. Schulz L.C. Ezashi T. Roberts R.M. Early onset preeclampsia in a model for human placental trophoblast.Proc. Natl. Acad. Sci. U. S. A. 2019; 116: 4336-4345Crossref PubMed Scopus (20) Google Scholar). However, whether bona fide trophoblast can be obtained from hPSCs has been a subject of intense debate (7Roberts R.M. Loh K.M. Amita M. Bernardo A.S. Adachi K. Alexenko A.P. Schust D.J. Schulz L.C. Telugu B.P.V.L. Ezashi T. Pedersen R.A. Differentiation of trophoblast cells from human embryonic stem cells: To be or not to be?.Reproduction (Cambridge, England). 2014; 147: D1-D12Crossref PubMed Scopus (45) Google Scholar). A rigorous head-to-head comparison between trophoblast derived from hPSCs and their in vivo counterparts has proven difficult owing to multiple reasons. Previous studies have used varying experimental protocols (8Roberts R.M. Ezashi T. Sheridan M.A. Yang Y. Specification of trophoblast from embryonic stem cells exposed to BMP4†.Biol. Reprod. 2018; 99: 212-224Crossref PubMed Scopus (22) Google Scholar); both primary placental samples and cultures of terminally differentiated trophoblast obtained from hPSCs exhibit heterogeneity and contain many cell types, and until recently self-renewing TS-like cells had not been derived from hPSCs (9Dong C. Beltcheva M. Gontarz P. Zhang B. Popli P. Fischer L.A. Khan S.A. Park K.-M. Yoon E.-J. Xing X. Kommagani R. Wang T. Solnica-Krezel L. Theunissen T.W. Derivation of trophoblast stem cells from naïve human pluripotent stem cells.eLife. 2020; 9: e52504Crossref PubMed Scopus (57) Google Scholar, 10Cinkornpumin J.K. Kwon S.Y. Guo Y. Hossain I. Sirois J. Russett C.S. Tseng H.W. Okae H. Arima T. Duchaine T.F. Liu W. Pastor W.A. Naive human embryonic stem cells can give rise to cells with a trophoblast-like transcriptome and Methylome.Stem Cell Rep. 2020; 15: 198-213Abstract Full Text Full Text PDF PubMed Scopus (33) Google Scholar, 11Li Z. Kurosawa O. Iwata H. Development of trophoblast cystic structures from human induced pluripotent stem cells in limited-area cell culture.Biochem. Biophysical Res. Commun. 2018; 505: 671-676Crossref PubMed Scopus (5) Google Scholar, 12Gao X. Nowak-Imialek M. Chen X. Chen D. Herrmann D. Ruan D. Chen A.C.H. Eckersley-Maslin M.A. Ahmad S. Lee Y.L. Kobayashi T. Ryan D. Zhong J. Zhu J. Wu J. et al.Establishment of porcine and human expanded potential stem cells.Nat. Cell Biol. 2019; 21: 687-699Crossref PubMed Scopus (120) Google Scholar). In this study, we report the derivation and maintenance of two distinct trophectoderm lineage stem cell types from hPSCs, specifically human embryonic stem cells (hESCs) and hiPSCs, in chemically defined culture conditions. The first is a CDX2- stem cell that is comparable with TS cells derived from early-gestation placental samples and similar to the villous CTB. The second is a CDX2+ cell type with distinct cell culture requirements, and differences in gene expression and differentiation, relative to CDX2- stem cells. Critically, the isolation of self-renewing stem cell populations allowed a direct comparison of placenta-derived TS cells with TS cells from hPSCs; genome-wide transcriptomic analysis and functional differentiation assays demonstrate very high similarity between placenta- and hPSC-derived CDX2- TS cells. The routine derivation of TS cells from hPSCs will provide powerful tools for mechanistic studies on normal and pathological early trophoblast development. Media formulations in previous studies on trophoblast differentiation of hESCs included components such as knockout serum replacement (KSR) or bovine serum albumin (BSA) that act as carriers for lipids. Albumin-associated lipids have been implicated in activation of G-protein–coupled receptor–mediated signaling (13Yu F.-X. Zhao B. Panupinthu N. Jewell J.L. Lian I. Wang L.H. Zhao J. Yuan H. Tumaneng K. Li H. Fu X.-D. Mills G.B. Guan K.-L. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling.Cell. 2012; 150: 780-791Abstract Full Text Full Text PDF PubMed Scopus (974) Google Scholar, 14Mendelson K. Evans T. Hla T. Sphingosine 1-phosphate signalling.Development (Cambridge, England). 2014; 141: 5-9Crossref PubMed Scopus (165) Google Scholar). For instance, the phospholipid sphingosine-1 phosphate (S1P) present in KSR can activate YAP signaling. YAP plays a critical role in specification of the trophectoderm in mouse (15Yagi R. Kohn M.J. Karavanova I. Kaneko K.J. Vullhorst D. DePamphilis M.L. Buonanno A. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development.Development (Cambridge, England). 2007; 134: 3827-3836Crossref PubMed Scopus (353) Google Scholar, 16Knott J.G. Paul S. Transcriptional regulators of the trophoblast lineage in mammals with hemochorial placentation.Reproduction (Cambridge, England). 2014; 148: R121-R136Crossref PubMed Scopus (40) Google Scholar, 17Nishioka N. Yamamoto S. Kiyonari H. Sato H. Sawada A. Ota M. Nakao K. Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos.Mech. Dev. 2008; 125: 270-283Crossref PubMed Scopus (331) Google Scholar), as well as human trophoblast development (18Saha B. Ganguly A. Home P. Bhattacharya B. Ray S. Ghosh A. Rumi M.A.K. Marsh C. French V. Gunewardena S. Paul S. TEAD4 ensures postimplantation development by promoting trophoblast self-renewal: An implication in early human pregnancy loss.Proc. Natl. Acad. Sci. 2020; 117: 202002449Crossref Scopus (20) Google Scholar, 19Meinhardt G. Haider S. Kunihs V. Saleh L. Pollheimer J. Fiala C. Hetey S. Feher Z. Szilagyi A. Than N.G. Knöfler M. Pivotal role of the transcriptional co-activator YAP in trophoblast stemness of the developing human placenta.Proc. Natl. Acad. Sci. U. S. A. 2020; 117: 13562-13570Crossref PubMed Scopus (25) Google Scholar). We investigated the use of S1P in the context of trophoblast differentiation of hESCs under chemically defined culture conditions, by modifying our previous protocol that utilized KSR (20Sarkar P. Randall S.M. Collier T.S. Nero A. Russell T.A. Muddiman D.C. Rao B.M. Activin/nodal signaling Switches the terminal fate of human embryonic stem cell-derived trophoblasts.J. Biol. Chem. 2015; 290: 8834-8848Abstract Full Text Full Text PDF PubMed Scopus (15) Google Scholar, 21Sarkar P. Mischler A. Randall S.M. Collier T.S. Dorman K.F. Boggess K.A. Muddiman D.C. Rao B.M. Identification of epigenetic factor proteins expressed in human embryonic stem cell-derived trophoblasts and in human placental trophoblasts.J. Proteome Res. 2016; 15: 2433-2444Crossref PubMed Scopus (6) Google Scholar). H1 and H9 hESCs cultured in E8 medium were differentiated for 6 days in E7 medium (E8 without transforming growth factor-beta1 [TGFβ1]) supplemented with S1P, by treatment with BMP4 and the activin/nodal inhibitor SB431542 (Fig. 1A). Under these conditions, we observed upregulation of the trophectoderm marker CDX2 and the CTB marker ELF5 (Fig. S1, A and B). Upregulation of TBX4 was observed after 6 days. However, overall there were no significant changes in markers associated with neural or mesodermal differentiation after 6 days suggesting that differentiation to these lineages did not occur (Fig. S1, A and B). Immunofluorescence analysis at day 6 confirmed expression of the pan-trophoblast marker KRT7, and CTB markers P63 and GATA3; expression of CDX2 was not observed (Figs. 1B and S1C). The putative CTB cells obtained at day 6 were investigated for their ability to differentiate to EVTs and STB, using protocols similar to those previously employed (20Sarkar P. Randall S.M. Collier T.S. Nero A. Russell T.A. Muddiman D.C. Rao B.M. Activin/nodal signaling Switches the terminal fate of human embryonic stem cell-derived trophoblasts.J. Biol. Chem. 2015; 290: 8834-8848Abstract Full Text Full Text PDF PubMed Scopus (15) Google Scholar). We observed formation of mesenchymal cells from epithelial cells over a 6-day period when passaged into E8 medium supplemented with epidermal growth factor (EGF) and SB431542. Immunofluorescence analysis showed expression of KRT7 and the EVT markers VE-Cadherin and HLA-G (Figs. 1C, S1D). Alternatively, passaging CTB-like cells in E6 medium (E8 without TGFβ1 and fibroblast growth factor-2 [FGF2]) supplemented with activin and EGF resulted in the formation of KRT7+ multinucleate cells expressing the STB markers hCG and syncytin over an 8-day period (Figs. 1D, S1E). Removal of S1P from the medium during hESC differentiation to CTB-like cells abolished the formation of EVTs that express HLA-G and VE-Cadherin (Figs. 1E, S2A) under identical differentiation conditions (Fig. 1A). Differentiation to STB also did not occur in the absence of S1P, as evidenced by lack of expression of syncytin and KRT7 (Figs. 1F, S2B). Also, downregulation of the trophectoderm marker CDX2 and upregulation of transcripts of neural and mesoderm markers was observed in cells after 6 days of differentiation, upon removal of S1P (Fig. S2C). Taken together these results show that CTB-like cells, similar to those in previous studies utilizing more complex culture conditions (20Sarkar P. Randall S.M. Collier T.S. Nero A. Russell T.A. Muddiman D.C. Rao B.M. Activin/nodal signaling Switches the terminal fate of human embryonic stem cell-derived trophoblasts.J. Biol. Chem. 2015; 290: 8834-8848Abstract Full Text Full Text PDF PubMed Scopus (15) Google Scholar), can be obtained by differentiation of hESCs in a chemically defined medium containing S1P. Furthermore, addition of exogenous S1P is necessary for hESC differentiation to trophoblast in our chemically defined culture medium. Rho GTPase signaling, downstream of G-protein–coupled receptors activated by S1P, has been implicated in nuclear localization of YAP (22Ohgushi M. Minaguchi M. Sasai Y. Rho-signaling-directed YAP/TAZ activity Underlies the long-term Survival and Expansion of human embryonic stem cells.Cell stem cell. 2015; 17: 448-461Abstract Full Text Full Text PDF PubMed Scopus (100) Google Scholar, 23Mo J.-S. Yu F.-X. Gong R. Brown J.H. Guan K.-L. Regulation of the Hippo-YAP pathway by protease-activated receptors (PARs).Genes Dev. 2012; 26: 2138-2143Crossref PubMed Scopus (195) Google Scholar). Both Rho/RhoA associated kinase (ROCK) and nuclear YAP play a critical role in trophectoderm specification in the mouse (24Nishioka N. Inoue K. Adachi K. Kiyonari H. Ota M. Ralston A. Yabuta N. Hirahara S. Stephenson R.O. Ogonuki N. Makita R. Kurihara H. Morin-Kensicki E.M. Nojima H. Rossant J. et al.The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass.Dev. Cel. 2009; 16: 398-410Abstract Full Text Full Text PDF PubMed Scopus (657) Google Scholar, 25Kono K. Tamashiro D.A.A. Alarcon V.B. Inhibition of RHO-ROCK signaling enhances ICM and suppresses TE characteristics through activation of Hippo signaling in the mouse blastocyst.Dev. Biol. 2014; 394: 142-155Crossref PubMed Scopus (79) Google Scholar). Therefore, we investigated the role of Rho/ROCK signaling and YAP in trophoblast differentiation of hESCs. The Rho/ROCK inhibitor Y-27632 was included during differentiation of hESCs to CTB-like cells and subsequent differentiation to EVT and STB to investigate the role of Rho/ROCK signaling. Under these conditions, HLA-G expression was observed in cells obtained from H9 hESCs; however, VE-Cadherin expression was weak and observed in only a few cells (Fig. S3A). On the other hand, expression of EVT markers was not observed in cells derived from H1 hESCs. In addition, presence of ROCK inhibition abolished STB formation, as shown by the lack of expression of syncytin and KRT7 (Fig. S3B). To investigate the role of YAP signaling in CTB formation from hESCs, we used an hESC cell line (H9) that expresses an inducible shRNA against YAP (H9-YAP-ishRNA) or a scrambled shRNA control (26Hsiao C. Lampe M. Nillasithanukroh S. Han W. Lian X. Palecek S.P. Human pluripotent stem cell culture density modulates YAP signaling.Biotechnol. J. 2016; 11: 662-675Crossref PubMed Scopus (26) Google Scholar). YAP knockdown abolished differentiation to EVT and STB, as evidenced by lack of expression of the relevant markers. It is notable that high cell death was observed (Fig. S3, A and B). Gene expression analysis revealed a significant reduction in ELF5 upon YAP knockdown, relative to the scrambled shRNA control (Fig. S3C). Significant downregulation of the mesodermal genes TBX4 and LMO2 was observed, whereas T was upregulated, in H9-YAP-ishRNA, relative to the scrambled control. Taken together, these results show that Rho/ROCK signaling and YAP are necessary for differentiation of hESCs to functional CTB that can give rise to both EVTs and STB, in our chemically defined culture medium. S1P acts through both receptor-mediated and receptor-independent pathways (14Mendelson K. Evans T. Hla T. Sphingosine 1-phosphate signalling.Development (Cambridge, England). 2014; 141: 5-9Crossref PubMed Scopus (165) Google Scholar, 27Maceyka M. Harikumar K.B. Milstien S. Spiegel S. Sphingosine-1-phosphate signaling and its role in disease.Trends Cell Biol. 2012; 22: 50-60Abstract Full Text Full Text PDF PubMed Scopus (695) Google Scholar). To investigate the specific mechanism of S1P action during hESC differentiation to trophoblast, we replaced S1P with D-erythro-dihydrospingosine-1-phosphate (dhS1P) in our protocol. dhS1P acts as an agonist for the S1P receptors (S1PRs) but does not mediate an intracellular effect (28Van Brocklyn J.R. Lee M.-J. Menzeleev R. Olivera A. Edsall L. Cuvillier O. Thomas D.M. Coopman P.J.P. Thangada S. Liu C.H. Hla T. Spiegel S. Dual actions of sphingosine-1-phosphate: Extracellular through the G i -coupled receptor Edg-1 and intracellular to regulate proliferation and Survival.J. Cell Biol. 1998; 142: 229-240Crossref PubMed Scopus (444) Google Scholar). Replacing S1P with dhS1P yielded similar results—CTB-like cells showed expression of CDX2, GATA3, P63, and TEAD4 (Figs. 2A and S4A). Upon further differentiation as previously described (Fig. 1A), STB expressing KRT7 and hCG, and EVT expressing HLA-G and VE-Cadherin were obtained (Fig. 2, B and C; Fig. S4, B and C). These results suggest that S1PR signaling mediates the effect of exogenous S1P during hESC differentiation to trophoblast in our chemically defined medium. S1P acts extracellularly through S1PR1-5 (14Mendelson K. Evans T. Hla T. Sphingosine 1-phosphate signalling.Development (Cambridge, England). 2014; 141: 5-9Crossref PubMed Scopus (165) Google Scholar, 27Maceyka M. Harikumar K.B. Milstien S. Spiegel S. Sphingosine-1-phosphate signaling and its role in disease.Trends Cell Biol. 2012; 22: 50-60Abstract Full Text Full Text PDF PubMed Scopus (695) Google Scholar); however, TBs have been shown to only express S1PR1-3 (29Johnstone E.D. Chan G. Sibley C.P. Davidge S.T. Lowen B. Guilbert L.J. Sphingosine-1-phosphate inhibition of placental trophoblast differentiation through a G(i)-coupled receptor response.J. lipid Res. 2005; 46: 1833-1839Abstract Full Text Full Text PDF PubMed Scopus (34) Google Scholar). We further used selective chemical agonists for S1PR1-3—CYM5442 hydrochloride, CYM5520, and CYM5541, respectively—to replace S1P in differentiation protocols previously discussed. Expression of CDX2, GATA3, P63, and TEAD4 was observed in CTB-like cells for all three agonists (Figs. 2A and S4A). Similarly, use of each agonist resulted in expression of the EVT markers HLA-G and VE-Cadherin and formation of multinucleate STB expressing KRT7 and hCG (Fig. 2, B and C; Fig. S4, B and C). However, we observed some variability between the agonists (Fig. S5). For instance, use of the S1PR2 agonist resulted in strong cytoplasmic expression of P63 and high heterogeneity in staining at day 6 relative to the other agonists. Formation of large multinucleated STB was more pronounced when the S1PR2 or S1PR3 agonists were used, as compared with the S1PR1 agonist. On the other hand, the S1PR1 and S1PR3 agonists enhanced the formation of mesenchymal EVTs, relative to the S1PR2 agonist. Taken together, our results further confirmed that S1PR signaling mediates effects of exogenous S1P during trophoblast differentiation of hESCs in our culture system. Since our qualitative observations showed that use of the S1PR3 agonist resulted in expression of CTB markers, and both multinucleate STB and mesenchymal EVTs could be obtained when the S1PR3 agonist was used, we chose the S1PR3 agonist for subsequent studies. We investigated whether CTB-like cells obtained by treatment of hESCs with BMP4 and SB431542 in E7 medium supplemented with the S1PR3 agonist CYM5541 for 6 days could be passaged and maintained under conditions used for culture of blastocyst- and placenta-derived primary TS cells (5Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. Kabayama Y. Suyama M. Sasaki H. Arima T. Derivation of human trophoblast stem cells.Cell stem cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (233) Google Scholar). Upon plating in trophoblast stem cell medium (TSCM) developed by Okae et al. (5Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. Kabayama Y. Suyama M. Sasaki H. Arima T. Derivation of human trophoblast stem cells.Cell stem cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (233) Google Scholar), hESC-derived CTB-like cells underwent differentiation and epithelial colonies could not be retained after a single passage. CDX2 expression is upregulated significantly in as little as 2 days after initiation of hESC differentiation but decreases by day 6 (Fig. S1, A and B). In addition, previous studies have reported differentiation of hESCs to CDX2+/p63+ cells upon treatment with BMP for 4 days (30Horii M. Li Y. Wakeland A.K. Pizzo D.P. Nelson K.K. Sabatini K. Laurent L.C. Liu Y. Parast M.M. Human pluripotent stem cells as a model of trophoblast differentiation in both normal development and disease.Proc. Natl. Acad. Sci. United States America. 2016; 113: E3882-E3891Crossref PubMed Scopus (66) Google Scholar). Therefore, we explored the use of a shorter differentiation step for obtaining CTB-like cells (Fig. 3A). After 3 days of differentiation, H9 and H1 hESCs expressed nuclear CDX2, P63, and TEAD4 uniformly (Fig. 3B). However, by day 6 most differentiated H1 and H9 hESCs lose expression of CDX2 (Fig. 3C). Quantitative image analysis showed that nearly all cells are CDX2+ at day 3, in contrast to CTB-like cells at day 6. Of note, use of a 6-day protocol resulted in a significantly reduced fraction of CDX2+ cells in the case of H1 hESCs in comparison with the 3-day protocol; on the other hand, a significant fraction of H9 cells retained CDX2+ at day 6 (Fig. 3D). Transcriptome analysis using RNA sequencing identified 291 genes with significantly higher expression levels and 330 genes with significantly lower expression levels in day 3 differentiated hESCs versus undifferentiated hESCs (Tables S1 and S2).Expression of other trophectoderm-associated markers such as HAND1, GATA3, and TFAP2A, in addition to CDX2, was upregulated in differentiated hESCs at day 3, whereas expression of pluripotency-associated NANOG was downregulated. Gene set enrichment analysis of differentially expressed genes identified 567 and 202 gene ontology (GO) categories (of 9996 queried categories) associated with higher and lower gene expression in day 3 differentiated cells versus undifferentiated hESCs, respectively (Tables S3 and S4). Consistent with differentiation to epithelial trophoblast, genes associated with the GO terms for epithelium development, epithelial cell proliferation, and epithelial cell differentiation were upregulated in day 3 differentiated hESCs. CDX2+ cells at day 3 were passaged into a chemically defined medium containing four major components (denoted TM4), the S1PR3 agonist CYM5541, the GSK3β inhibitor CHIR99021, the TGFβ inhibitor A83-01, and FGF10. CHIR99021 and A83-01 are components of TSCM used for culture of primary TS cells; FGF10 was included because FGFR2b signaling is active in blastocyst- and placenta-derived TS cells and the early placenta (5Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. Kabayama Y. Suyama M. Sasaki H. Arima T. Derivation of human trophoblast stem cells.Cell stem cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (233) Google Scholar). Cells in TM4 could be maintained as epithelial colonies for 30+ passages over the course of 5 months. In TM4 medium, cells derived from H9 and H1 hESCs retained expression of the trophoblast markers CDX2, TFAP2C, YAP, TEAD4, and GATA3 (Figs. 3E and S6) (15Yagi R. Kohn M.J. Karavanova I. Kaneko K.J. Vullhorst D. DePamphilis M.L. Buonanno A. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development.Development (Cambridge, England). 2007; 134: 3827-3836Crossref PubMed Scopus (353) Google Scholar, 17Nishioka N. Yamamoto S. Kiyonari H. Sato H. Sawada A. Ota M. Nakao K. Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos.Mech. Dev. 2008; 125: 270-283Crossref PubMed Scopus (331) Google Scholar, 31Choi I. Carey T.S. Wilson C.A. Knott J.G. Transcription factor AP-2γ is a core regulator of tight junction biogenesis and cavity formation during mouse early embryogenesis.Development (Cambridge, England). 2012; 139: 4623-4632Crossref PubMed Scopus (40) Google Scholar, 32Home P. Saha B. Ray S. Dutta D. Gunewardena S. Yoo B. Pal A. Vivian J.L. Larson M. Petroff M. Gallagher P.G. Schulz V.P. White K.L. Golos T.G. Behr B. et al.Altered subcellular localization of transcription factor TEAD4 regulates first mammalian cell lineage commitment.Proc. Natl. Acad. Sci. U. S. A. 2012; 109: 7362-7367Crossref PubMed Scopus (113) Google Scholar, 33Ralston A. Cox B.J. Nishioka N. Sasaki H. Chea E. Rugg-Gunn P. Guo G. Robson P. Draper J.S. Rossant J. Gata3 regulates trophoblast development downstream of Tead4 and in parallel to Cdx2.Development (Cambridge, England). 2010; 137: 395-403Crossref PubMed Scopus (299) Google Scholar, 34Niwa H. Toyooka Y. Shimosato D. Strumpf D. Takahashi K. Yagi R. Rossant J. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation.Cell. 2005; 123: 917-929Abstract Full Text Full Text PDF PubMed Scopus (854) Google Scholar). In addition, cells expressed the pan-trophoblast marker KRT7 and low levels of P63. Of note, CDX2 expression has been strongly associated with the trophectoderm and is lost once placental villi are formed (30Horii M. Li Y. Wakeland A.K. Pizzo D.P. Nelson K.K. Sabatini K. Laurent L.C. Liu Y. Parast M.M. Human pluripotent stem cells as a model of trophoblast differentiation in both normal development and disease.Proc. Natl. Acad. Sci. United States America. 2016; 113: E3882-E3891Crossref PubMed Scopus (66) Google Scholar, 35Hemberger M. Udayashankar R. Tesar P. Moore H. Burton G.J. ELF5-enforced transcriptional networks define an epigenetically regulated trophoblast stem cell compartment in the human placenta.Hum. Mol. Genet. 2010; 19: 2456-2467Crossref PubMed Scopus (126) Google Scholar, 36Blakeley P. Fogarty N.M.E. del Valle I. Wamaitha S.E. Hu T.X. Elder K. Snell P. Christie L. Robson P. Niakan K.K. Defining the three cell lineages of the human blastocyst by single-cell RNA-seq.Development. 2015; 142: 3151-3165Crossref PubMed Scopus (262) Google Scholar, 37Knöfler M. Haider S. Saleh L. Pollheimer J. Gamage T.K.J.B. James J. Human placenta and trophoblast development: Key molecular mechanisms and model systems.Cell Mol. Life Sci. 2019; 76: 3479-3496Crossref PubMed Scopus (144) Google Scholar). To indicate that these cells are derived from hPSCs, and to distinguish these cells from TS cells that do not express CDX2, these cells are denoted as hPSC-TSCDX2 cells. We further evaluated the differentiation potential of hPSC-TSCDX2 cells using same protocols as those used by Okae et al. for differentiation of primary TS cells to EVTs and STB (5Okae H. Toh H. Sato T. Hiura H. Takahashi S. Shirane K. Kabayama Y. Suyama M. Sasaki H. Arima T. Derivation of human trophoblast stem cells.Cell stem cell. 2018; 22: 50-63.e6Abstract Full Text Full Text PDF PubMed Scopus (233) Google Scholar). Cells were able to form multinucleate STB that expressed hCG and KRT7 (Fig. 3F). However, upon EVT treatment, cells did not form mesenchymal elongated cells but acquired a flattened morphology. Upon passage, cells showed no HLA-G and minimal VE-Cadherin expression (Fig. 3G). Furthermore, cells maintained an epithelial flattened morp}, journal={JOURNAL OF BIOLOGICAL CHEMISTRY}, publisher={Elsevier BV}, author={Mischler, Adam and Karakis, Victoria and Mahinthakumar, Jessica and Carberry, Celeste K. and San Miguel, Adriana and Rager, Julia E. and Fry, Rebecca C. and Rao, Balaji M.}, year={2021} } @article{mischler_karakis_san miguel_rao_2019, title={DERIVATION OF HUMAN TROPHOBLAST STEM CELLS FROM HUMAN PLURIPOTENT STEM CELLS}, volume={83}, ISSN={["1532-3102"]}, DOI={10.1016/j.placenta.2019.06.193}, abstractNote={Conventional soil maps contain valuable knowledge on soil–environment relationships. Such knowledge can be extracted for use when updating conventional soil maps with improved environmental data. Existing methods take all polygons of the same map unit on a map as a whole to extract the soil–environment relationship. Such approach ignores the difference in the environmental conditions represented by individual soil polygons of the same map unit. This paper proposes a method of mining soil–environment relationships from individual soil polygons to update conventional soil maps. The proposed method consists of three major steps. Firstly, the soil–environment relationships represented by each individual polygon on a conventional soil map are extracted in the form of frequency distribution curves for the involved environmental covariates. Secondly, for each environmental covariate, these frequency distribution curves from individual polygons of the same soil map unit are synthesized to form the overall soil–environment relationship for that soil map unit across the mapped area. And lastly, the extracted soil–environment relationships are applied to updating the conventional soil map with new, improved environmental data by adopting a soil land inference model (SoLIM) framework. This study applied the proposed method to updating a conventional soil map of the Raffelson watershed in La Crosse County, Wisconsin, United States. The result from the proposed method was compared with that from the previous method of taking all polygons within the same soil map unit on a map as a whole. Evaluation results with independent soil samples showed that the proposed method exhibited better performance and produced higher accuracy.}, journal={PLACENTA}, author={Mischler, Adam and Karakis, Victoria and San Miguel, Adriana and Rao, Balaji}, year={2019}, month={Aug}, pages={E59–E59} }