@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.}, 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} } @article{tiruthani_mischler_ahmed_mahinthakumar_haugh_rao_2019, title={Design and evaluation of engineered protein biosensors for live-cell imaging of EGFR phosphorylation}, volume={12}, ISSN={["1937-9145"]}, DOI={10.1126/scisignal.aap7584}, abstractNote={Engineered biosensors accurately report the kinetics of EGFR activation.}, number={584}, journal={SCIENCE SIGNALING}, author={Tiruthani, Karthik and Mischler, Adam and Ahmed, Shoeb and Mahinthakumar, Jessica and Haugh, Jason M. and Rao, Balaji M.}, year={2019}, month={Jun} } @article{cruz-teran_tiruthani_mischler_rao_2017, title={Inefficient Ribosomal Skipping Enables Simultaneous Secretion and Display of Proteins in Saccharomyces cerevisiae}, volume={6}, ISSN={["2161-5063"]}, DOI={10.1021/acssynbio.7b00144}, abstractNote={The need for recombinant expression of soluble protein slows the validation of engineered proteins isolated from combinatorial libraries and limits the number of protein variants evaluated. To overcome this bottleneck, we describe a system for simultaneous cell surface display and soluble secretion of proteins in Saccharomyces cerevisiae based on inefficient ribosomal skipping. Ribosomal skipping mediated by "self-cleaving" 2A peptides produces two proteins from a single open reading frame. Incorporation of the F2A peptide sequence-with ∼50% efficiency of ribosomal skipping-between the protein of interest and the yeast cell wall protein Aga2 results in simultaneous expression of both the solubly secreted protein and the protein-Aga2 fusion that is tethered to the yeast cell surface. We show that binding proteins derived from the Sso7d scaffold and the homodimeric enzyme glucose oxidase can be simultaneously secreted solubly and expressed as yeast cell surface fusions using the F2A-based system. Furthermore, a combinatorial library of Sso7d mutants can be screened to isolate binders with higher affinity for a model target (lysozyme), and the pool of higher affinity binders can be characterized in soluble form. Significantly, we show that both N- and C-terminal fusions to Aga2 can be simultaneously secreted solubly and displayed on the cell surface; this is particularly advantageous because protein functionality can be affected by the specific position of Aga2 in the protein fusion. We expect that the F2A-based yeast surface display and secretion system will be a useful tool for protein engineering and enable efficient characterization of individual clones isolated from combinatorial libraries.}, number={11}, journal={ACS SYNTHETIC BIOLOGY}, author={Cruz-Teran, Carlos A. and Tiruthani, Karthik and Mischler, Adam and Rao, Balaji M.}, year={2017}, month={Nov}, pages={2096–2107} } @article{sarkar_mischler_randall_collier_dorman_boggess_muddiman_rao_2016, title={Identification of Epigenetic Factor Proteins Expressed in Human Embryonic Stem Cell-Derived Trophoblasts and in Human Placental Trophoblasts}, volume={15}, ISSN={["1535-3907"]}, DOI={10.1021/acs.jproteome.5b01118}, abstractNote={Human embryonic stem cells (hESCs) have been used to derive trophoblasts through differentiation in vitro. Intriguingly, mouse ESCs are prevented from differentiation to trophoblasts by certain epigenetic factor proteins such as Dnmt1, thus necessitating the study of epigenetic factor proteins during hESC differentiation to trophoblasts. We used stable isotope labeling by amino acids in cell culture and quantitative proteomics to study changes in the nuclear proteome during hESC differentiation to trophoblasts and identified changes in the expression of 30 epigenetic factor proteins. Importantly, the DNA methyltransferases DNMT1, DNMT3A, and DNMT3B were downregulated. Additionally, we hypothesized that nuclear proteomics of hESC-derived trophoblasts may be used for screening epigenetic factor proteins expressed by primary trophoblasts in human placental tissue. Accordingly, we conducted immunohistochemistry analysis of six epigenetic factor proteins identified from hESC-derived trophoblasts-DNMT1, DNMT3B, BAF155, BAF60A, BAF57, and ING5-in 6-9 week human placentas. Indeed, expression of these proteins was largely, though not fully, consistent with that observed in 6-9 week placental trophoblasts. Our results support the use of hESC-derived trophoblasts as a model for placental trophoblasts, which will enable further investigation of epigenetic factors involved in human trophoblast development.}, number={8}, journal={JOURNAL OF PROTEOME RESEARCH}, author={Sarkar, Prasenjit and Mischler, Adam and Randall, Shan M. and Collier, Timothy S. and Dorman, Karen F. and Boggess, Kim A. and Muddiman, David C. and Rao, Balaji M.}, year={2016}, month={Aug}, pages={2433–2444} }