1
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. Dev Biol 2025; 524:105-115. [PMID: 40349907 DOI: 10.1016/j.ydbio.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
During the first cell fate decision in mammalian embryos, the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell and nuclear shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
2
|
Zhang W, An S, Hou S, He X, Xiang J, Yan H, Liu X, Dong L, Wang X, Yang Y. Generation of transient totipotent blastomere-like stem cells by short-term high-dose Pladienolide B treatment. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1337-1351. [PMID: 40024996 DOI: 10.1007/s11427-024-2774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/10/2024] [Indexed: 03/04/2025]
Abstract
As an alternative model for studying the dynamic process of early mammalian embryonic development, much progress has been made in using mouse embryonic stem cells (mESCs) to generate embryo-like structures, especially by modifying the starting cells. A previous study has demonstrated that totipotent blastomere-like cells (TBLCs) can be obtained by continuous treatment of mESCs with a low-dose splicing inhibitor, Pladienolide B (PlaB). However, these totipotent mESCs have limited proliferative capacity. Here, we report that short-term high-dose PlaB treatment can also induce mESCs to acquire totipotency. This treatment equips this novel type of stem cells with the ability to self-organize into blastoids and recapitulate key preimplantation developmental processes. Therefore, the stem cells are termed transient totipotent blastomere-like stem cells (tTBLCs). Transcriptome analysis showed that tTBLC blastoids bore similarities to mouse E3.5 blastocysts, E4.5 blastocysts, and TBLC blastoids. Additionally, we found that tTBLC blastoids could develop beyond the implantation stage, forming egg-cylinder-like structures both in vitro and in vivo. In summary, our research provides an alternative rapid and convenient method to generate the starting cells capable of developing into blastoids, which have immense application in various fields, not only in the basic study of early mouse embryogenesis but also in high-throughput drug screening.
Collapse
Affiliation(s)
- Wenyi Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shiyu An
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shuyue Hou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xingsi He
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jinfeng Xiang
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, 210004, China
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Institute, Nanjing, 210004, China
| | - Huanyu Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaorui Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Lingling Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China.
- Department of Prenatal Diagnosis of the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210036, China.
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China.
- Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, China.
| |
Collapse
|
3
|
Jeong M, Han D, Bhetariya P, Welling DB, Stojkovic M, Stankovic KM. NF2 is Essential for Human Endoderm Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410909. [PMID: 39921490 PMCID: PMC12061267 DOI: 10.1002/advs.202410909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/18/2024] [Indexed: 02/10/2025]
Abstract
Vertebrate embryogenesis requires the precisely timed specification of 3 germ cell layers- ectoderm, mesoderm, and endoderm- which give rise to tissues and organs in the developing organism. The tumor suppressor gene NF2, moesin-ezrin-radixin like (MERLIN) tumor suppressor (Nf2) is expressed in all 3 germ layers during mouse development and its homozygous deletion causes embryonic lethality. People with heterozygous NF2 mutations typically develop Schwann cell tumors, especially vestibular schwannoma, but the specific role of NF2 in human embryonic development is unclear. Here, human induced pluripotent stem cells (hiPSCs) are used to demonstrate that NF2 is essential for endoderm specification and formation in humans. Although endoderm differentiation is not impaired in hiPSCs with heterozygous NF2 mutation, NF2 knockout (NF2-/-) abolished the capacity to form endoderm in vitro, confirmed by loss of expression of endoderm-related genes and proteins, or teratomas in vivo. This defect is mediated by the nuclear translocation of yes-associated protein 1 (YAP1), a transcription co-activator regulating lineage fate via the Hippo pathway and subsequent YAP1-mediated shutdown of Activin/Nodal signaling. Endoderm formation can be rescued via YAP1 knockdown or forced re-expression of NF2 in NF2-/- cells. Taken together, the essential role of NF2 during endoderm specification in human embryogenesis as a regulator of YAP1 is reported.
Collapse
Affiliation(s)
- Minjin Jeong
- Department of Otolaryngology‐Head and Neck SurgeryStanford University School of MedicineStanfordCA94305USA
- Department of Otolaryngology‐Head and Neck SurgeryMassachusetts Eye and Ear and Harvard Medical SchoolBostonMA02114USA
| | - Dongjun Han
- Department of Otolaryngology‐Head and Neck SurgeryStanford University School of MedicineStanfordCA94305USA
- Department of Otolaryngology‐Head and Neck SurgeryMassachusetts Eye and Ear and Harvard Medical SchoolBostonMA02114USA
| | - Preetida Bhetariya
- Bioinformatics CoreHarvard T.H. Chan School of Public HealthBostonMA02115USA
| | - D. Bradley Welling
- Department of Otolaryngology‐Head and Neck SurgeryMassachusetts Eye and Ear and Harvard Medical SchoolBostonMA02114USA
| | - Miodrag Stojkovic
- Department of Otolaryngology‐Head and Neck SurgeryMassachusetts Eye and Ear and Harvard Medical SchoolBostonMA02114USA
| | - Konstantina M. Stankovic
- Department of Otolaryngology‐Head and Neck SurgeryStanford University School of MedicineStanfordCA94305USA
- Department of Otolaryngology‐Head and Neck SurgeryMassachusetts Eye and Ear and Harvard Medical SchoolBostonMA02114USA
- Department of NeurosurgeryStanford University School of MedicineStanfordCA94304USA
- Wu Tsai Neurosciences InstituteStanford UniversityStanfordCA94305USA
| |
Collapse
|
4
|
McGinnis AJ, Cull ME, Peterson NT, Tang MK, Natale BV, Natale DRC. Exploring the differentiation potential of Eomes POS mouse trophoblast cells in mid-gestation. Dev Biol 2025; 521:75-84. [PMID: 39922418 DOI: 10.1016/j.ydbio.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Mouse trophoblast stem (mTS) cells can be derived from the blastocyst or extraembryonic ectoderm as late as embryonic day (E) 6.5 and when cultured in vitro, can differentiate to all trophoblast subtypes of the mature placenta. Expression of the T-box transcription factor, Eomes, is required for the maintenance of, and used to identify mTS cells. During development, Eomes is restricted to the ExE and, by E7.5, to the chorion, after which its expression declines. The placental junctional zone and labyrinth layers are thought to develop exclusively from the ectoplacental cone and chorion, respectively. While it is well established that mTS cells express Eomes in vitro, it is unknown if Eomes-positive (EomesPOS) trophoblast that reside in the chorion after E6.5 are restricted in their developmental potential to the labyrinth layer in vivo. This study utilized a lineage tracing technique to evaluate the in vivo differentiation of EomesPOS trophoblast. Using an Ai6 reporter mouse crossed with a tamoxifen-inducible Eomes-Cre-ERT2 mouse, Cre was activated from E7.5 to E9.5, permanently marking all EomesPOS trophoblast and daughter cells with the ZsGreen fluorescent protein. This approach was complemented with immunofluorescence staining to assess how the EomesPOS trophoblast had contributed to the differentiated trophoblast population within the placenta by E17.5. Importantly, the results show that daughter cells of EomesPOS trophoblast in which Cre was activated, contributed to both placental layers; specifically, spongiotrophoblast and glycogen trophoblast within the junctional zone and syncytiotrophoblast and sinusoidal trophoblast giant cells within the labyrinth. This confirms that EomesPOS trophoblast maintain the capacity to contribute to both placental layers in vivo and do so after E7.5. This study expands our understanding of trophoblast differentiation in vivo and may prove useful in assessing how EomesPOS trophoblast contribute placental development later in gestation and in the context of placental pathology, where Eomes expression has been reported.
Collapse
Affiliation(s)
- Avery J McGinnis
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada
| | - Megan E Cull
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada
| | - Nichole T Peterson
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada
| | - Matthew K Tang
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada
| | - Bryony V Natale
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada
| | - David R C Natale
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
5
|
Dalal K, McAnany C, Weilert M, McKinney MC, Krueger S, Zeitlinger J. Interpreting regulatory mechanisms of Hippo signaling through a deep learning sequence model. CELL GENOMICS 2025; 5:100821. [PMID: 40174587 PMCID: PMC12008814 DOI: 10.1016/j.xgen.2025.100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/23/2024] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
Signaling pathway components are well studied, but how they mediate cell-type-specific transcription responses is an unresolved problem. Using the Hippo pathway in mouse trophoblast stem cells as a model, we show that the DNA binding of signaling effectors is driven by cell-type-specific sequence rules that can be learned genome wide by deep learning models. Through model interpretation and experimental validation, we show that motifs for the cell-type-specific transcription factor TFAP2C enhance TEAD4/YAP1 binding in a nucleosome-range and distance-dependent manner, driving synergistic enhancer activation. We also discovered that Tead double motifs are widespread, highly active canonical response elements. Molecular dynamics simulations suggest that TEAD4 binds them cooperatively through surprisingly labile protein-protein interactions that depend on the DNA template. These results show that the response to signaling pathways is encoded in the cis-regulatory sequences and that interpreting the rules reveals insights into the mechanisms by which signaling effectors influence cell-type-specific enhancer activity.
Collapse
Affiliation(s)
- Khyati Dalal
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Charles McAnany
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Melanie Weilert
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Sabrina Krueger
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Julia Zeitlinger
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
6
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640568. [PMID: 40060487 PMCID: PMC11888467 DOI: 10.1101/2025.02.27.640568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
During the first cell fate decision in mammalian embryos the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
7
|
Luo Y, An C, Zhong K, Zhou P, Li D, Liu H, Guo Q, Wei W, Pan H, Min Z, Li R, Yu Y, Fan Y. Exploring the impacts of senescence on implantation and early embryonic development using totipotent cell-derived blastoids. J Adv Res 2025; 68:115-129. [PMID: 38402947 PMCID: PMC11785586 DOI: 10.1016/j.jare.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
INTRODUCTION Advanced maternal age is associated with reduced implantation and pregnancy rates, yet the underlying mechanisms remain poorly understood, and research models are limited. OBJECTIVES Here, we aim to elucidate the impacts of senescence on implantation ability by employing blastoids to construct a novel research model. METHODS We used a novel three-dimensional system with totipotent blastomere-like cells (TBLCs) to construct TBL-blastoids and established senescence-related embryo models derived from oxidative stress-induced TBLCs. RESULTS Morphological and transcriptomic analyses revealed that TBL-blastoids exhibited characteristic blastocyst morphology, cell lineages, and a higher consistency in developmental rate. TBL-blastoids demonstrated the ability to develop into postimplantation structures in vitro and successfully implanted into mouse uteri, inducing decidualization and forming embryonic tissues. Importantly, senescence impaired the implantation potential of TBL-blastoids, effectively mimicking the impaired implantation ability and reduced pregnancy rates associated with advanced age. Furthermore, analysis of differentially expressed genes (DEGs) in human homologous deciduae revealed enrichment in multiple fertility-related diseases and other complications of pregnancy. The genes implicated in these diseases and the common DEGs identified in the lineage-like cells of the two types of TBL-blastoids and deciduae may represent potential targets for addressing impaired implantation potential. CONCLUSION These results unveiled that TBL blastoids are an improved model for investigating implantation and early postimplantation, offering valuable insights into pregnancy-related disorders in women with advanced age and potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Yuxin Luo
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Chenrui An
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Ke Zhong
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Ping Zhou
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Dan Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wei
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hen Pan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Zheying Min
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
8
|
Wu J, Shao T, Tang Z, Liu G, Li Z, Shi Y, Kang Y, Zuo J, Zhao B, Hu G, Liu J, Ji W, Zhang L, Niu Y. Highly efficient construction of monkey blastoid capsules from aged somatic cells. Nat Commun 2025; 16:1130. [PMID: 39875393 PMCID: PMC11775175 DOI: 10.1038/s41467-025-56447-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Blastoids-blastocyst-like structures created in vitro-emerge as a valuable model for early embryonic development research. Non-human primates stem cell-derived blastoids are an ethically viable alternative to human counterparts, yet the low formation efficiency of monkey blastoid cavities, typically below 30%, has limited their utility. Prior research has predominantly utilized embryonic stem cells. In this work, we demonstrate the efficient generation of blastoids from induced pluripotent stem cells and somatic cell nuclear transfer embryonic stem cells derived from aged monkeys, achieving an 80% formation efficiency. We also introduce a hydrogel-based microfluidics platform for the scalable and reproducible production of size-adjustable, biodegradable blastoid capsules, providing a stable 3D structure and mechanical protection. This advancement in the high-efficiency, scalable production of monkey blastoid capsules from reprogrammed aged somatic cells significantly enhances the study of embryonic development and holds promise for regenerative medicine.
Collapse
Affiliation(s)
- Junmo Wu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Tianao Shao
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Zengli Tang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Gaojing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuoyao Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yuxi Shi
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiawei Zuo
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Bo Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guangyu Hu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiaqi Liu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Lei Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Southwest United Graduate School, Kunming, Yunnan, China.
| |
Collapse
|
9
|
Marikawa Y, Alarcon VB. Regulation of trophectoderm morphogenesis by small GTPase RHOA through HIPPO signaling-dependent and -independent mechanisms in mouse preimplantation development. Differentiation 2025; 141:100835. [PMID: 39874642 PMCID: PMC11790356 DOI: 10.1016/j.diff.2025.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/27/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025]
Abstract
The trophectoderm (TE) is the first tissue to differentiate during the preimplantation development of the mammalian embryo. It forms the outer layer of the blastocyst and is responsible for generating the blastocoel, a fluid-filled cavity whose expansion is essential for successful hatching and implantation. Here, we investigated the role of the small GTPase RHOA in the morphogenesis of the TE, particularly its relationship with HIPPO signaling, using mouse embryos as a model. Inhibition of RHOA resulted in the failure to form a blastocoel and significantly altered the expression of numerous genes. Transcriptomic analysis revealed that 330 genes were down-regulated and 168 genes were up-regulated by more than two-fold. Notably, 98.4% of these transcriptional changes were reversed by simultaneous inhibition of LATS kinases, indicating that the transcriptional influence of RHOA is primarily mediated through HIPPO signaling. Many of the down-regulated genes are involved in critical processes of TE morphogenesis, such as apical-basal cell polarization, tight junction formation, and sodium and water transport, suggesting that RHOA supports TE development by enhancing the expression of morphogenesis-related genes through HIPPO signaling, specifically via TEAD transcription factors. However, RHOA inhibition also disrupted apical-basal polarity and tight junctions, effects that were not restored by LATS inhibition, pointing to additional HIPPO signaling-independent mechanisms by which RHOA controls TE morphogenesis. Furthermore, RHOA inhibition impaired cell viability at the late blastocyst stage, with partial rescue observed upon LATS inhibition, suggesting that RHOA maintains cell survival through both HIPPO signaling-dependent and -independent pathways. A deeper knowledge of the molecular mechanisms governing TE morphogenesis, including blastocoel expansion and cell viability, could significantly advance assisted reproductive technologies aimed at producing healthy blastocysts.
Collapse
Affiliation(s)
- Yusuke Marikawa
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA
| | - Vernadeth B Alarcon
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA.
| |
Collapse
|
10
|
Hirono N, Hashimoto M, Shimojo H, Sasaki H. Fate specification triggers a positive feedback loop of TEAD-YAP and NANOG to promote epiblast formation in preimplantation embryos. Development 2025; 152:dev203091. [PMID: 39629521 DOI: 10.1242/dev.203091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
In preimplantation embryos, epiblast (EPI) fate specification from the inner cell mass is controlled by the segregation of NANOG and GATA6 expression. TEAD-YAP interaction is activated during EPI formation and is required for pluripotency factor expression. These events occur asynchronously with similar timing during EPI formation, and their relationship remains elusive. Here, we examined the relationship between NANOG-GATA6 and TEAD-YAP. The nuclear accumulation of YAP takes place only in EPI-specified cells, and a positive feedback loop operates between NANOG and TEAD-YAP. The effects of TEAD-YAP on SOX2 upregulation in EPI-specified cells are likely indirect. EPI fate specification also alters the response of Nanog, Sox2 and Cdx2 to TEAD-YAP. These results suggest that EPI-fate specification alters the transcriptional network from the morula-like to the EPI-specified state and activates TEAD-YAP to trigger a positive feedback loop with NANOG, which stabilizes the EPI fate. The coordinated occurrence of these processes in individual cells likely supports proper EPI formation under the condition of asynchronous EPI-fate specification.
Collapse
Affiliation(s)
- Naoki Hirono
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masakazu Hashimoto
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
- Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama 332-0012, Japan
| | - Hiromi Shimojo
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Karami N, Taei A, Eftekhari-Yazdi P, Hassani F. Signaling pathway regulators in preimplantation embryos. J Mol Histol 2024; 56:57. [PMID: 39729177 DOI: 10.1007/s10735-024-10338-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Embryonic development during the preimplantation stages is highly sensitive and critically dependent on the reception of signaling cues. The precise coordination of diverse pathways and signaling factors is essential for successful embryonic progression. Even minor disruptions in these factors can result in physiological dysfunction, fetal malformations, or embryonic arrest. This issue is particularly evident in assisted reproductive technologies, such as in vitro fertilization, where embryonic arrest is frequently observed. A detailed understanding of these pathways enhances insight into the fundamental mechanisms underlying cellular processes and their contributions to embryonic development. The significance of elucidating signaling pathways and their regulatory factors in preimplantation development cannot be overstated. The application of this knowledge in laboratory settings has the potential to support strategies for modeling developmental stages and diseases, drug screening, therapeutic discovery, and reducing embryonic arrest. Furthermore, using various factors, small molecules, and pharmacological agents can enable the development or optimization of culture media for enhanced embryonic viability. While numerous pathways influence preimplantation development, this study examines several critical signaling pathways in this contex.
Collapse
Affiliation(s)
- Narges Karami
- MSc., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran.
| |
Collapse
|
12
|
Wong KW, Zeng Y, Tay E, Teo JHJ, Cipta NO, Hamashima K, Yi Y, Liu H, Warrier T, Le MTN, Ng SC, Li QJ, Li H, Loh YH. Nuclear receptor-SINE B1 network modulates expanded pluripotency in blastoids and blastocysts. Nat Commun 2024; 15:10011. [PMID: 39562549 PMCID: PMC11577042 DOI: 10.1038/s41467-024-54381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Embryonic stem cells possess the remarkable ability to self-organize into blastocyst-like structures upon induction. These stem cell-based embryo models serve as invaluable platforms for studying embryogenesis and therapeutic developments. Nevertheless, the specific intrinsic regulators that govern this potential for blastoid formation remain unknown. Here we demonstrate an intrinsic program that plays a crucial role in both blastoids and blastocysts across multiple species. We first establish metrics for grading the resemblance of blastoids to mouse blastocysts, and identify the differential activation of gene regulons involved in lineage specification among various blastoid grades. Notably, abrogation of nuclear receptor subfamily 1, group H, member 2 (Nr1h2) drastically reduces blastoid formation. Nr1h2 activation alone is sufficient to rewire conventional ESC into a distinct pluripotency state, enabling them to form blastoids with enhanced implantation capacity in the uterus and contribute to both embryonic and extraembryonic lineages in vivo. Through integrative multi-omics analyses, we uncover the broad regulatory role of Nr1h2 in the transcriptome, chromatin accessibility and epigenome, targeting genes associated with embryonic lineage and the transposable element SINE-B1. The Nr1h2-centred intrinsic program governs and drives the development of both blastoids and early embryos.
Collapse
Grants
- R03 OD038392 NIH HHS
- U19 AG074879 NIA NIH HHS
- R03 OD034496 NIH HHS
- P30 CA015083 NCI NIH HHS
- P30 DK084567 NIDDK NIH HHS
- P50 CA136393 NCI NIH HHS
- National Research Foundation, Singapore (NRF) Investigatorship award [NRFI2018- 02]; National Medical Research Council [NMRC/OFIRG21nov-0088]; Singapore Food Story (SFS) R&D Programme [W22W3D0007]; A*STAR Biomedical Research Council, Central Research Fund, Use-Inspired Basic Research (CRF UIBR); Competitive Research Programme (CRP) [NRF-CRP29-2022-0005]; Industry Alignment Fund - Prepositioning (IAF-PP) [H23J2a0095, H23J2a0097].
- NMRC grant MOH-000937-00 and A*STAR grant C210812003
- M.T.N.L. was supported by the Industry Alignment Fund - Prepositioning (IAF-PP) [H23J2a0097].
- H.L. was supported by grants from the Mayo Clinic Center for Biomedical Discovery, Center for Individualized Medicine, the Mayo Clinic Comprehensive Cancer Center (NIH; P30CA015083), the Mayo Clinic Center for Cell Signaling in Gastroenterology (NIH: P30DK084567), the Mayo Clinic Nutrition Obesity Research Program, the Glenn Foundation for Medical Research, the Eric & Wendy Schmidt Fund for AI Research & Innovation and the National Institutes of Health (NIH; U19AG74879, P50CA136393, R03OD038392).
Collapse
Affiliation(s)
- Ka Wai Wong
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yingying Zeng
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Edison Tay
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Jia Hao Jackie Teo
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Nadia Omega Cipta
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Kiyofumi Hamashima
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yao Yi
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Haijun Liu
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Tushar Warrier
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Minh T N Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Soon Chye Ng
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Sincere Healthcare Group, Singapore, Republic of Singapore
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Yuin-Han Loh
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Department of Biological Sciences, National University of Singapore, Singapore, Republic of Singapore.
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Republic of Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
13
|
Pérez-Gómez A, González-Brusi L, Flores-Borobia I, Galiano-Cogolludo B, Lamas-Toranzo I, Hamze JG, Toledano-Díaz A, Santiago-Moreno J, Ramos-Ibeas P, Bermejo-Álvarez P. The role of TEAD4 in trophectoderm commitment and development is not conserved in non-rodent mammals. Development 2024; 151:dev202993. [PMID: 39171364 PMCID: PMC11463960 DOI: 10.1242/dev.202993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
The first lineage differentiation in mammals gives rise to the inner cell mass and the trophectoderm (TE). In mice, TEAD4 is a master regulator of TE commitment, as it regulates the expression of other TE-specific genes and its ablation prevents blastocyst formation, but its role in other mammals remains unclear. Herein, we have observed that TEAD4 ablation in two phylogenetically distant species (bovine and rabbit) does not impede TE differentiation, blastocyst formation and the expression of TE markers, such as GATA3 and CDX2, although a reduced number of cells in the inner cell mass was observed in bovine TEAD4 knockout (KO) blastocysts. Transcriptional analysis in bovine blastocysts revealed no major transcriptional effect of the ablation, although the expression of hypoblast and Hippo signalling-related genes tended to be decreased in KO embryos. Experiments were conducted in the bovine model to determine whether TEAD4 was required for post-hatching development. TEAD4 KO spherical conceptuses showed normal development of the embryonic disc and TE, but hypoblast migration rate was reduced. At later stages of development (tubular conceptuses), no differences were observed between KO and wild-type conceptuses.
Collapse
|
14
|
Russell SJ, Zhao C, Biondic S, Menezes K, Hagemann-Jensen M, Librach CL, Petropoulos S. An atlas of small non-coding RNAs in human preimplantation development. Nat Commun 2024; 15:8634. [PMID: 39367016 PMCID: PMC11452719 DOI: 10.1038/s41467-024-52943-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Understanding the molecular circuitries that govern early embryogenesis is important, yet our knowledge of these in human preimplantation development remains limited. Small non-coding RNAs (sncRNAs) can regulate gene expression and thus impact blastocyst formation, however, the expression of specific biotypes and their dynamics during preimplantation development remains unknown. Here we identify the abundance of and kinetics of piRNA, rRNA, snoRNA, tRNA, and miRNA from embryonic day (E)3-7 and isolate specific miRNAs and snoRNAs of particular importance in blastocyst formation and pluripotency. These sncRNAs correspond to specific genomic hotspots: an enrichment of the chromosome 19 miRNA cluster (C19MC) in the trophectoderm (TE), and the chromosome 14 miRNA cluster (C14MC) and MEG8-related snoRNAs in the inner cell mass (ICM), which may serve as 'master regulators' of potency and lineage. Additionally, we observe a developmental transition with 21 isomiRs and in tRNA fragment (tRF) codon usage and identify two novel miRNAs. Our analysis provides a comprehensive measure of sncRNA biotypes and their corresponding dynamics throughout human preimplantation development, providing an extensive resource. Better understanding the sncRNA regulatory programmes in human embryogenesis will inform strategies to improve embryo development and outcomes of assisted reproductive technologies. We anticipate broad usage of our data as a resource for studies aimed at understanding embryogenesis, optimising stem cell-based models, assisted reproductive technology, and stem cell biology.
Collapse
MESH Headings
- Humans
- RNA, Small Untranslated/genetics
- RNA, Small Untranslated/metabolism
- Embryonic Development/genetics
- Blastocyst/metabolism
- Gene Expression Regulation, Developmental
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- Female
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/genetics
- Chromosomes, Human, Pair 19/genetics
- RNA, Small Nucleolar/genetics
- RNA, Small Nucleolar/metabolism
Collapse
Affiliation(s)
| | - Cheng Zhao
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Savana Biondic
- Faculty of Medicine, Molecular Biology Program, Université de Montréal, Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montréal, Canada
| | | | | | - Clifford L Librach
- CReATe Fertility Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sophie Petropoulos
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
- Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden.
- Faculty of Medicine, Molecular Biology Program, Université de Montréal, Montréal, QC, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montréal, Canada.
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Faculty of Medicine, Département de Médecine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
15
|
Festuccia N, Vandormael-Pournin S, Chervova A, Geiselmann A, Langa-Vives F, Coux RX, Gonzalez I, Collet GG, Cohen-Tannoudji M, Navarro P. Nr5a2 is dispensable for zygotic genome activation but essential for morula development. Science 2024; 386:eadg7325. [PMID: 39361745 DOI: 10.1126/science.adg7325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/10/2024] [Accepted: 08/05/2024] [Indexed: 10/05/2024]
Abstract
Early embryogenesis is driven by transcription factors (TFs) that first activate the zygotic genome and then specify the lineages constituting the blastocyst. Although the TFs specifying the blastocyst's lineages are well characterized, those playing earlier roles remain poorly defined. Using mouse models of the TF Nr5a2, we show that Nr5a2-/- embryos arrest at the early morula stage and exhibit altered lineage specification, frequent mitotic failure, and substantial chromosome segregation defects. Although NR5A2 plays a minor but measurable role during zygotic genome activation, it predominantly acts as a master regulator at the eight-cell stage, controlling expression of lineage-specifying TFs and genes involved in mitosis, telomere maintenance, and DNA repair. We conclude that NR5A2 coordinates proliferation, genome stability, and lineage specification to ensure correct morula development.
Collapse
Affiliation(s)
- Nicola Festuccia
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Sandrine Vandormael-Pournin
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Almira Chervova
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Anna Geiselmann
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
- Sorbonne Université, Complexité du Vivant, 75005 Paris, France
| | | | - Rémi-Xavier Coux
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Inma Gonzalez
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Guillaume Giraud Collet
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
- Université Paris Cité, BioSPC, 75013 Paris, France
| | - Michel Cohen-Tannoudji
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| | - Pablo Navarro
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3738, Epigenomics, Proliferation, and the Identity of Cells Unit, 75015 Paris, France
| |
Collapse
|
16
|
Wang S, Shao D, Gao X, Zhao P, Kong F, Deng J, Yang L, Shang W, Sun Y, Fu Z. TEAD transcription factor family emerges as a promising therapeutic target for oral squamous cell carcinoma. Front Immunol 2024; 15:1480701. [PMID: 39430767 PMCID: PMC11486717 DOI: 10.3389/fimmu.2024.1480701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
The treatment of oral squamous cell carcinoma (OSCC) remains a significant difficulty, as there has been no improvement in survival rates over the past fifty years. Hence, exploration and confirmation of new dependable treatment targets and biomarkers is imperative for OSCC therapy. TEAD transcription factors are crucial for integrating and coordinating multiple signaling pathways that are essential for embryonic development, organ formation, and tissue homeostasis. In addition, by attaching to coactivators, TEAD modifies the expression of genes such as Cyr61, Myc, and connective tissue growth factor, hence facilitating tumor progression. Therefore, TEAD is regarded as an effective predictive biomarker due to its significant connection with clinical parameters in several malignant tumors, including OSCC. The efficacy of existing drugs that specifically target TEAD has demonstrated encouraging outcomes, indicating its potential as an optimal target for OSCC treatment. This review provides an overview of current targeted therapy strategies for OSCC by highlighting the transcription mechanism and involvement of TEAD in oncogenic signaling pathways. Finally, the feasibility of utilizing TEAD as an innovative approach to address OSCC and its potential clinical applications were analyzed and discussed.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
- Department of Stomatology, Medical College of Qingdao Huanghai University, Qingdao, China
| | - Dan Shao
- Department of Oral and Maxillofacial Surgery, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiaoyan Gao
- Department of Quality Inspection, Traditional Chinese Medical Hospital of Huangdao District, Qingdao, China
| | - Peng Zhao
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
| | - Fanzhi Kong
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
| | - Jiawei Deng
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
| | - Lianzhu Yang
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
| | - Wei Shang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaping Sun
- Department of Stomatology, Qingdao West Coast New District Central Hospital, Qingdao, China
| | - Zhiguang Fu
- Department of Tumor Radiotherapy, Air Force Medical Center, People's Liberation Army of China (PLA), Beijing, China
| |
Collapse
|
17
|
Pfeffer PL. The first lineage determination in mammals. Dev Biol 2024; 513:12-30. [PMID: 38761966 DOI: 10.1016/j.ydbio.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/15/2024] [Accepted: 05/16/2024] [Indexed: 05/20/2024]
Abstract
This review describes in detail the morphological, cytoskeletal and gene expression events leading to the gene regulatory network bifurcation point of trophoblast and inner cell mass cells in a variety of mammalian preimplantation embryos. The interrelated processes of compaction and polarity establishment are discussed in terms of how they affect YAP/WWTR activity and the location and fate of cells. Comparisons between mouse, human, cattle, pig and rabbit embryos suggest a conserved role for YAP/WWTR signalling in trophoblast induction in eutherian animals though the mechanisms for, and timing of, YAP/WWTR activation differs among species. Downstream targets show further differences, with the trophoblast marker GATA3 being a direct target in all examined mammals, while CDX2-positive and SOX2-negative regulation varies.
Collapse
Affiliation(s)
- Peter L Pfeffer
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
18
|
Skory RM. Revisiting trophectoderm-inner cell mass lineage segregation in the mammalian preimplantation embryo. Hum Reprod 2024; 39:1889-1898. [PMID: 38926157 PMCID: PMC12102071 DOI: 10.1093/humrep/deae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
In the first days of life, cells of the mammalian embryo segregate into two distinct lineages, trophectoderm and inner cell mass. Unlike nonmammalian species, mammalian development does not proceed from predetermined factors in the oocyte. Rather, asymmetries arise de novo in the early embryo incorporating cues from cell position, contractility, polarity, and cell-cell contacts. Molecular heterogeneities, including transcripts and non-coding RNAs, have now been characterized as early as the 2-cell stage. However, it's debated whether these early heterogeneities bias cells toward one fate or the other or whether lineage identity arises stochastically at the 16-cell stage. This review summarizes what is known about early blastomere asymmetries and our understanding of lineage allocation in the context of historical models. Preimplantation development is reviewed coupled with what is known about changes in morphology, contractility, and transcription factor networks. The addition of single-cell atlases of human embryos has begun to reveal key differences between human and mouse, including the timing of events and core transcription factors. Furthermore, the recent generation of blastoid models will provide valuable tools to test and understand fate determinants. Lastly, new techniques are reviewed, which may better synthesize existing knowledge with emerging data sets and reconcile models with the regulative capacity unique to the mammalian embryo.
Collapse
Affiliation(s)
- Robin M Skory
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Zhu M, Meglicki M, Lamba A, Wang P, Royer C, Turner K, Jauhar MA, Jones C, Child T, Coward K, Na J, Zernicka-Goetz M. Tead4 and Tfap2c generate bipotency and a bistable switch in totipotent embryos to promote robust lineage diversification. Nat Struct Mol Biol 2024; 31:964-976. [PMID: 38789684 PMCID: PMC11189297 DOI: 10.1038/s41594-024-01311-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/09/2024] [Indexed: 05/26/2024]
Abstract
The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.
Collapse
Affiliation(s)
- Meng Zhu
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Maciej Meglicki
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Adiyant Lamba
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Peizhe Wang
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Christophe Royer
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Karen Turner
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, UK
| | - Muhammad Abdullah Jauhar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tim Child
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jie Na
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
20
|
Driscoll CS, Kim J, Knott JG. The explosive discovery of TNT in early mouse embryos. Nat Struct Mol Biol 2024; 31:852-855. [PMID: 38789683 DOI: 10.1038/s41594-024-01304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Affiliation(s)
- Chad S Driscoll
- Developmental Epigenetics Laboratory, Department of Animal Science, Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Jaehwan Kim
- Developmental Epigenetics Laboratory, Department of Animal Science, Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Jason G Knott
- Developmental Epigenetics Laboratory, Department of Animal Science, Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
21
|
Kim HJ, Choi Y, Lee Y, Hwangbo M, Kim J. OTUD6A orchestrates complex modulation of TEAD4-mediated transcriptional programs. FEBS Lett 2024; 598:1045-1060. [PMID: 38594215 DOI: 10.1002/1873-3468.14861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 04/11/2024]
Abstract
TEAD transcription factors play a central role in the Hippo signaling pathway. In this study, we focused on transcriptional enhancer factor TEF-3 (TEAD4), exploring its regulation by the deubiquitinase OTU domain-containing protein 6A (OTUD6A). We identified OTUD6A as a TEAD4-interacting deubiquitinase, positively influencing TEAD-driven transcription without altering TEAD4 stability. Structural analyses revealed specific interaction domains: the N-terminal domain of OTUD6A and the YAP-binding domain of TEAD4. Functional assays demonstrated the positive impact of OTUD6A on the transcription of YAP-TEAD target genes. Despite no impact on TEAD4 nuclear localization, OTUD6A selectively modulated nuclear interactions, enhancing YAP-TEAD4 complex formation while suppressing VGLL4 (transcription cofactor vestigial-like protein 4)-TEAD4 interaction. Critically, OTUD6A facilitated YAP-TEAD4 complex binding to target gene promoters. Our study unveils the regulatory landscape of OTUD6A on TEAD4, providing insights into diseases regulated by YAP-TEAD complexes.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yunsik Choi
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yuri Lee
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Mi Hwangbo
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Jongchan Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| |
Collapse
|
22
|
Azagury M, Buganim Y. Unlocking trophectoderm mysteries: In vivo and in vitro perspectives on human and mouse trophoblast fate induction. Dev Cell 2024; 59:941-960. [PMID: 38653193 DOI: 10.1016/j.devcel.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/10/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
In recent years, the pursuit of inducing the trophoblast stem cell (TSC) state has gained prominence as a compelling research objective, illuminating the establishment of the trophoblast lineage and unlocking insights into early embryogenesis. In this review, we examine how advancements in diverse technologies, including in vivo time course transcriptomics, cellular reprogramming to TSC state, chemical induction of totipotent stem-cell-like state, and stem-cell-based embryo-like structures, have enriched our insights into the intricate molecular mechanisms and signaling pathways that define the mouse and human trophectoderm/TSC states. We delve into disparities between mouse and human trophectoderm/TSC fate establishment, with a special emphasis on the intriguing role of pluripotency in this context. Additionally, we re-evaluate recent findings concerning the potential of totipotent-stem-like cells and embryo-like structures to fully manifest the trophectoderm/trophoblast lineage's capabilities. Lastly, we briefly discuss the potential applications of induced TSCs in pregnancy-related disease modeling.
Collapse
Affiliation(s)
- Meir Azagury
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
23
|
Dupont C. A comprehensive review: synergizing stem cell and embryonic development knowledge in mouse and human integrated stem cell-based embryo models. Front Cell Dev Biol 2024; 12:1386739. [PMID: 38715920 PMCID: PMC11074781 DOI: 10.3389/fcell.2024.1386739] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mammalian stem cell-based embryo models have emerged as innovative tools for investigating early embryogenesis in both mice and primates. They not only reduce the need for sacrificing mice but also overcome ethical limitations associated with human embryo research. Furthermore, they provide a platform to address scientific questions that are otherwise challenging to explore in vivo. The usefulness of a stem cell-based embryo model depends on its fidelity in replicating development, efficiency and reproducibility; all essential for addressing biological queries in a quantitative manner, enabling statistical analysis. Achieving such fidelity and efficiency requires robust systems that demand extensive optimization efforts. A profound understanding of pre- and post-implantation development, cellular plasticity, lineage specification, and existing models is imperative for making informed decisions in constructing these models. This review aims to highlight essential differences in embryo development and stem cell biology between mice and humans, assess how these variances influence the formation of partially and fully integrated stem cell models, and identify critical challenges in the field.
Collapse
Affiliation(s)
- Cathérine Dupont
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
24
|
Nita A, Moroishi T. Hippo pathway in cell-cell communication: emerging roles in development and regeneration. Inflamm Regen 2024; 44:18. [PMID: 38566194 PMCID: PMC10986044 DOI: 10.1186/s41232-024-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
The Hippo pathway is a central regulator of tissue growth that has been widely studied in mammalian organ development, regeneration, and cancer biology. Although previous studies have convincingly revealed its cell-autonomous functions in controlling cell fate, such as cell proliferation, survival, and differentiation, accumulating evidence in recent years has revealed its non-cell-autonomous functions. This pathway regulates cell-cell communication through direct interactions, soluble factors, extracellular vesicles, and the extracellular matrix, providing a range of options for controlling diverse biological processes. Consequently, the Hippo pathway not only dictates the fate of individual cells but also triggers multicellular responses involving both tissue-resident cells and infiltrating immune cells. Here, we have highlighted the recent understanding of the molecular mechanisms by which the Hippo pathway controls cell-cell communication and discuss its importance in tissue homeostasis, especially in development and regeneration.
Collapse
Affiliation(s)
- Akihiro Nita
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
25
|
Chea S, Kreger J, Lopez-Burks ME, MacLean AL, Lander AD, Calof AL. Gastrulation-stage gene expression in Nipbl+/- mouse embryos foreshadows the development of syndromic birth defects. SCIENCE ADVANCES 2024; 10:eadl4239. [PMID: 38507484 PMCID: PMC10954218 DOI: 10.1126/sciadv.adl4239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
In animal models, Nipbl deficiency phenocopies gene expression changes and birth defects seen in Cornelia de Lange syndrome, the most common cause of which is Nipbl haploinsufficiency. Previous studies in Nipbl+/- mice suggested that heart development is abnormal as soon as cardiogenic tissue is formed. To investigate this, we performed single-cell RNA sequencing on wild-type and Nipbl+/- mouse embryos at gastrulation and early cardiac crescent stages. Nipbl+/- embryos had fewer mesoderm cells than wild-type and altered proportions of mesodermal cell subpopulations. These findings were associated with underexpression of genes implicated in driving specific mesodermal lineages. In addition, Nanog was found to be overexpressed in all germ layers, and many gene expression changes observed in Nipbl+/- embryos could be attributed to Nanog overexpression. These findings establish a link between Nipbl deficiency, Nanog overexpression, and gene expression dysregulation/lineage misallocation, which ultimately manifest as birth defects in Nipbl+/- animals and Cornelia de Lange syndrome.
Collapse
Affiliation(s)
- Stephenson Chea
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Jesse Kreger
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Martha E. Lopez-Burks
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Arthur D. Lander
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Anne L. Calof
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
26
|
Rashidiani S, Mamo G, Farkas B, Szabadi A, Farkas B, Uszkai V, Császár A, Brandt B, Kovács K, Pap M, Rauch TA. Integrative Epigenetic and Molecular Analysis Reveals a Novel Promoter for a New Isoform of the Transcription Factor TEAD4. Int J Mol Sci 2024; 25:2223. [PMID: 38396900 PMCID: PMC10888684 DOI: 10.3390/ijms25042223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/07/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
TEAD4 is a transcription factor that plays a crucial role in the Hippo pathway by regulating the expression of genes related to proliferation and apoptosis. It is also involved in the maintenance and differentiation of the trophectoderm during pre- and post-implantation embryonic development. An alternative promoter for the TEAD4 gene was identified through epigenetic profile analysis, and a new transcript from the intronic region of TEAD4 was discovered using the 5'RACE method. The transcript of the novel promoter encodes a TEAD4 isoform (TEAD4-ΔN) that lacks the DNA-binding domain but retains the C-terminal protein-protein interaction domain. Gene expression studies, including end-point PCR and Western blotting, showed that full-length TEAD4 was present in all investigated tissues. However, TEAD4-ΔN was only detectable in certain cell types. The TEAD4-ΔN promoter is conserved throughout evolution and demonstrates transcriptional activity in transient-expression experiments. Our study reveals that TEAD4 interacts with the alternative promoter and increases the expression of the truncated isoform. DNA methylation plays a crucial function in the restricted expression of the TEAD4-ΔN isoform in specific tissues, including the umbilical cord and the placenta. The data presented indicate that the DNA-methylation status of the TEAD4-ΔN promoter plays a critical role in regulating organ size, cancer development, and placenta differentiation.
Collapse
Affiliation(s)
- Shima Rashidiani
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary; (S.R.); (G.M.); (B.F.); (A.S.)
| | - Gizaw Mamo
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary; (S.R.); (G.M.); (B.F.); (A.S.)
| | - Benjámin Farkas
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary; (S.R.); (G.M.); (B.F.); (A.S.)
| | - András Szabadi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary; (S.R.); (G.M.); (B.F.); (A.S.)
- Department of Dentistry, Oral and Maxillofacial Surgery, Medical School, University of Pécs, 7623 Pécs, Hungary
| | - Bálint Farkas
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary; (B.F.); (V.U.); (A.C.)
- National Laboratory of Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Veronika Uszkai
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary; (B.F.); (V.U.); (A.C.)
| | - András Császár
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary; (B.F.); (V.U.); (A.C.)
| | - Barbara Brandt
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, 7624 Pécs, Hungary (M.P.)
| | - Kálmán Kovács
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary; (B.F.); (V.U.); (A.C.)
- National Laboratory of Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Marianna Pap
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, 7624 Pécs, Hungary (M.P.)
| | - Tibor A. Rauch
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary; (S.R.); (G.M.); (B.F.); (A.S.)
| |
Collapse
|
27
|
Gao Y, Han W, Dong R, Wei S, Chen L, Gu Z, Liu Y, Guo W, Yan F. Efficient Reprogramming of Mouse Embryonic Stem Cells into Trophoblast Stem-like Cells via Lats Kinase Inhibition. BIOLOGY 2024; 13:71. [PMID: 38392290 PMCID: PMC10886645 DOI: 10.3390/biology13020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024]
Abstract
Mouse zygotes undergo multiple rounds of cell division, resulting in the formation of preimplantation blastocysts comprising three lineages: trophectoderm (TE), epiblast (EPI), and primitive endoderm (PrE). Cell fate determination plays a crucial role in establishing a healthy pregnancy. The initial separation of lineages gives rise to TE and inner cell mass (ICM), from which trophoblast stem cells (TSC) and embryonic stem cells (ESC) can be derived in vitro. Studying lineage differentiation is greatly facilitated by the clear functional distinction between TSC and ESC. However, transitioning between these two types of cells naturally poses challenges. In this study, we demonstrate that inhibiting LATS kinase promotes the conversion of ICM to TE and also effectively reprograms ESC into stable, self-renewing TS-like cells (TSLC). Compared to TSC, TSLC exhibits similar molecular properties, including the high expression of marker genes such as Cdx2, Eomes, and Tfap2c, as well as hypomethylation of their promoters. Importantly, TSLC not only displays the ability to differentiate into mature trophoblast cells in vitro but also participates in placenta formation in vivo. These findings highlight the efficient reprogramming of ESCs into TSLCs using a small molecular inducer, which provides a new reference for understanding the regulatory network between ESCs and TSCs.
Collapse
Affiliation(s)
- Yake Gao
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
- Reproductive Medicine Center, Wuhan Women's and Children's Medical Care Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenrui Han
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Rui Dong
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Shu Wei
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Lu Chen
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Zhaolei Gu
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yiming Liu
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Wei Guo
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Fang Yan
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| |
Collapse
|
28
|
Yang Y, Jia W, Luo Z, Li Y, Liu H, Fu L, Li J, Jiang Y, Lai J, Li H, Saeed BJ, Zou Y, Lv Y, Wu L, Zhou T, Shan Y, Liu C, Lai Y, Liu L, Hutchins AP, Esteban MA, Mazid MA, Li W. VGLL1 cooperates with TEAD4 to control human trophectoderm lineage specification. Nat Commun 2024; 15:583. [PMID: 38233381 PMCID: PMC10794710 DOI: 10.1038/s41467-024-44780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
In contrast to rodents, the mechanisms underlying human trophectoderm and early placenta specification are understudied due to ethical barriers and the scarcity of embryos. Recent reports have shown that human pluripotent stem cells (PSCs) can differentiate into trophectoderm (TE)-like cells (TELCs) and trophoblast stem cells (TSCs), offering a valuable in vitro model to study early placenta specification. Here, we demonstrate that the VGLL1 (vestigial-like family member 1), which is highly expressed during human and non-human primate TE specification in vivo but is negligibly expressed in mouse, is a critical regulator of cell fate determination and self-renewal in human TELCs and TSCs derived from naïve PSCs. Mechanistically, VGLL1 partners with the transcription factor TEAD4 (TEA domain transcription factor 4) to regulate chromatin accessibility at target gene loci through histone acetylation and acts in cooperation with GATA3 and TFAP2C. Our work is relevant to understand primate early embryogenesis and how it differs from other mammalian species.
Collapse
Affiliation(s)
- Yueli Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenqi Jia
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiwei Luo
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Yunpan Li
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Hao Liu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Lixin Fu
- University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Jinxiu Li
- University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Yu Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junjian Lai
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
- BGI Research, Shenzhen, China
| | - Haiwei Li
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Babangida Jabir Saeed
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Yi Zou
- BGI Research, Shenzhen, China
| | - Yuan Lv
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
- BGI Research, Shenzhen, China
| | - Liang Wu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Ting Zhou
- Stem Cell Research Facility, Sloan Kettering Institute, New York, NY, USA
| | - Yongli Shan
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | | | - Yiwei Lai
- BGI Research, Shenzhen, China
- BGI Research, Hangzhou, China
| | - Longqi Liu
- BGI Research, Shenzhen, China
- BGI Research, Hangzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Andrew P Hutchins
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Miguel A Esteban
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China.
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China.
- BGI Research, Shenzhen, China.
- BGI Research, Hangzhou, China.
| | - Md Abdul Mazid
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China.
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China.
| | - Wenjuan Li
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, China.
- CAS Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China.
| |
Collapse
|
29
|
Yu T, Zhang C, Song W, Zhao X, Cheng Y, Liu J, Su J. Single-cell RNA-seq and single-cell bisulfite-sequencing reveal insights into yak preimplantation embryogenesis. J Biol Chem 2024; 300:105562. [PMID: 38097189 PMCID: PMC10821408 DOI: 10.1016/j.jbc.2023.105562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 01/13/2024] Open
Abstract
Extensive epigenetic reprogramming occurs during preimplantation embryonic development. However, the impact of DNA methylation in plateau yak preimplantation embryos and how epigenetic reprogramming contributes to transcriptional regulatory networks are unclear. In this study, we quantified gene expression and DNA methylation in oocytes and a series of yak embryos at different developmental stages and at single-cell resolution using single-cell bisulfite-sequencing and RNA-seq. We characterized embryonic genome activation and maternal transcript degradation and mapped epigenetic reprogramming events critical for embryonic development. Through cross-species transcriptome analysis, we identified 31 conserved maternal hub genes and 39 conserved zygotic hub genes, including SIN3A, PRC1, HDAC1/2, and HSPD1. Notably, by combining single-cell DNA methylation and transcriptome analysis, we identified 43 candidate methylation driver genes, such as AURKA, NUSAP1, CENPF, and PLK1, that may be associated with embryonic development. Finally, using functional approaches, we further determined that the epigenetic modifications associated with the histone deacetylases HDAC1/2 are essential for embryonic development and that the deubiquitinating enzyme USP7 may affect embryonic development by regulating DNA methylation. Our data represent an extensive resource on the transcriptional dynamics of yak embryonic development and DNA methylation remodeling, and provide new insights into strategies for the conservation of germplasm resources, as well as a better understanding of mammalian early embryonic development that can be applied to investigate the causes of early developmental disorders.
Collapse
Affiliation(s)
- Tong Yu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengtu Zhang
- Academician Zhang Yong Innovation Center, Xining Animal Disease Control Center, Xining, Qinghai, China
| | - Weijia Song
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinyi Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuyao Cheng
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Jianmin Su
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
30
|
Driskill JH, Pan D. Control of stem cell renewal and fate by YAP and TAZ. Nat Rev Mol Cell Biol 2023; 24:895-911. [PMID: 37626124 DOI: 10.1038/s41580-023-00644-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2023] [Indexed: 08/27/2023]
Abstract
Complex physiological processes control whether stem cells self-renew, differentiate or remain quiescent. Two decades of research have placed the Hippo pathway, a highly conserved kinase signalling cascade, and its downstream molecular effectors YAP and TAZ at the nexus of this decision. YAP and TAZ translate complex biological cues acting on stem cells - from mechanical forces to cellular metabolism - into genome-wide effects to mediate stem cell functions. While aberrant YAP/TAZ activity drives stem cell dysfunction in ageing, tumorigenesis and disease, therapeutic targeting of Hippo signalling and YAP/TAZ can boost stem cell activity to enhance regeneration. In this Review, we discuss how YAP/TAZ control the self-renewal, fate and plasticity of stem cells in different contexts, how dysregulation of YAP/TAZ in stem cells leads to disease, and how therapeutic modalities targeting YAP/TAZ may benefit regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Jordan H Driskill
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Lee J, Cai L, Kim M, Choi H, Oh D, Jawad A, Lee E, Hyun SH. Tetraploid embryo aggregation produces high-quality blastocysts with an increased trophectoderm in pigs. Front Cell Dev Biol 2023; 11:1239448. [PMID: 38033873 PMCID: PMC10687364 DOI: 10.3389/fcell.2023.1239448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Tetraploid complementation is an ideal method for demonstrating the differentiation potential of pluripotent stem cells. In this study, we selected the most efficient tetraploid production method for porcine embryos and investigated whether tetraploid blastomere aggregation could enhance the quality of tetraploid embryos. Three methods were investigated to produce tetraploid embryos: First, tetraploid embryos were produced using electro-fusion of two-cell stage parthenogenetic blastomere (FUTP). Second, somatic cell was injected into the mature oocyte and fused to produce tetraploid embryos. Third, oocytes were matured with Cytochalasin B (CB) for the late 22 h of in vitro maturation to inhibit the first polar body (PB1). Following that, non-PB1 oocytes were treated with CB for 4 h after parthenogenetic activation. There was no significant difference in the blastocyst development rate and tetraploid production rate of the embryos produced through the three methods. However, FUTP-derived blastocysts had a significantly lower percentage of apoptotic cells compared to other methods. The developmental competence of embryos, expression of trophectoderm cell marker genes, and distribution of YAP1 protein were investigated in tetraploid embryos produced using the FUTP method. The FUTP method most effectively prevented apoptosis during porcine tetraploid embryo formation. Tetraploid aggregation-derived blastocysts have a high proportion of trophectoderm with increased expression of the CDX2 mRNA and high YAP1 intensity. High-quality blastocysts derived from a tetraploid embryo aggregation can serve as suitable source material for testing the differentiation potential of pluripotent stem cells for blastocyst complementation in pigs.
Collapse
Affiliation(s)
- Joohyeong Lee
- Department of Companion Animal Industry, College of Healthcare and Biotechnology, Semyung University, Jecheon, Republic of Korea
| | - Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| | - Mirae Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Hyerin Choi
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjin Oh
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Ali Jawad
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Eunsong Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
32
|
Torre D, Francoeur NJ, Kalma Y, Gross Carmel I, Melo BS, Deikus G, Allette K, Flohr R, Fridrikh M, Vlachos K, Madrid K, Shah H, Wang YC, Sridhar SH, Smith ML, Eliyahu E, Azem F, Amir H, Mayshar Y, Marazzi I, Guccione E, Schadt E, Ben-Yosef D, Sebra R. Isoform-resolved transcriptome of the human preimplantation embryo. Nat Commun 2023; 14:6902. [PMID: 37903791 PMCID: PMC10616205 DOI: 10.1038/s41467-023-42558-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023] Open
Abstract
Human preimplantation development involves extensive remodeling of RNA expression and splicing. However, its transcriptome has been compiled using short-read sequencing data, which fails to capture most full-length mRNAs. Here, we generate an isoform-resolved transcriptome of early human development by performing long- and short-read RNA sequencing on 73 embryos spanning the zygote to blastocyst stages. We identify 110,212 unannotated isoforms transcribed from known genes, including highly conserved protein-coding loci and key developmental regulators. We further identify 17,964 isoforms from 5,239 unannotated genes, which are largely non-coding, primate-specific, and highly associated with transposable elements. These isoforms are widely supported by the integration of published multi-omics datasets, including single-cell 8CLC and blastoid studies. Alternative splicing and gene co-expression network analyses further reveal that embryonic genome activation is associated with splicing disruption and transient upregulation of gene modules. Together, these findings show that the human embryo transcriptome is far more complex than currently known, and will act as a valuable resource to empower future studies exploring development.
Collapse
Affiliation(s)
- Denis Torre
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Yael Kalma
- Fertility and IVF Institute, Tel-Aviv Sourasky Medical Center, Affiliated to Tel Aviv University, Tel Aviv, 64239, Israel
| | - Ilana Gross Carmel
- Fertility and IVF Institute, Tel-Aviv Sourasky Medical Center, Affiliated to Tel Aviv University, Tel Aviv, 64239, Israel
| | - Betsaida S Melo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gintaras Deikus
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kimaada Allette
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ron Flohr
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, 69978, Israel
- CORAL - Center Of Regeneration and Longevity, Tel-Aviv Sourasky Medical Center, Tel Aviv, 64239, Israel
| | - Maya Fridrikh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Kent Madrid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hardik Shah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shwetha H Sridhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Melissa L Smith
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, 40202, USA
| | - Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Foad Azem
- Fertility and IVF Institute, Tel-Aviv Sourasky Medical Center, Affiliated to Tel Aviv University, Tel Aviv, 64239, Israel
| | - Hadar Amir
- Fertility and IVF Institute, Tel-Aviv Sourasky Medical Center, Affiliated to Tel Aviv University, Tel Aviv, 64239, Israel
| | - Yoav Mayshar
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ivan Marazzi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Ernesto Guccione
- Center for OncoGenomics and Innovative Therapeutics (COGIT); Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dalit Ben-Yosef
- Fertility and IVF Institute, Tel-Aviv Sourasky Medical Center, Affiliated to Tel Aviv University, Tel Aviv, 64239, Israel.
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, 69978, Israel.
- CORAL - Center Of Regeneration and Longevity, Tel-Aviv Sourasky Medical Center, Tel Aviv, 64239, Israel.
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Wang M, Cheng R, He H, Han Z, Zhang Y, Wu Q. N 4-acetylcytidine of Nop2 mRNA is required for the transition of morula-to-blastocyst. Cell Mol Life Sci 2023; 80:307. [PMID: 37768430 PMCID: PMC11071819 DOI: 10.1007/s00018-023-04955-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) modification is crucial for mRNA stability and translation efficiency, yet the underlying function in mammalian preimplantation embryos remains unclear. Here, we characterized the ac4C modification landscape in mouse early embryos and found that the majority of embryos deficient in ac4C writer-NAT10 failed to develop into normal blastocysts. Through single-cell sequencing, RNA-seq, acetylated RNA immunoprecipitation combined with PCR (acRIP-PCR), and embryonic phenotype monitoring, Nop2 was screened as a target gene of Nat10. Mechanistically, Nat10 knockdown decreases the ac4C modification on Nop2 mRNA and reduces RNA and protein abundance by affecting the mRNA stability of Nop2. Then, depletion of NOP2 may inhibit the translation of transcription factor TEAD4, resulting in defective expression of the downstream lineage-specific gene Cdx2, and ultimately preventing blastomeres from undergoing the trophectoderm (TE) fate. However, exogenous Nop2 mRNA partially reverses this abnormal development. In conclusion, our findings demonstrate that defective ac4C modification of Nop2 mRNA hinders the morula-to-blastocyst transition by influencing the first cell fate decision in mice.
Collapse
Affiliation(s)
- Mengyun Wang
- Developmental Biology Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Rui Cheng
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Hongjuan He
- Developmental Biology Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhengbin Han
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yan Zhang
- Computational Biology Research Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| | - Qiong Wu
- Developmental Biology Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
34
|
Otsuka T, Shimojo H, Sasaki H. Daughter cells inherit YAP localization from mother cells in early preimplantation embryos. Dev Growth Differ 2023; 65:360-369. [PMID: 37309238 DOI: 10.1111/dgd.12870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
The first stage of cell differentiation during mouse development is the differentiation into the trophectoderm and inner cell mass, which occurs during the 8-32-cell stages of preimplantation embryos. This differentiation is regulated by the Hippo signaling pathway. At the 32-cell stage, embryos establish a position-dependent distribution of the Hippo pathway coactivator, Yes-associated protein 1 (YAP, encoded by Yap1). The outer and inner cells showed nuclear and cytoplasmic localization of YAP, respectively. However, the process by which embryos establish position-dependent YAP localization remains elusive. Here, we established a YAP-reporter mouse line, Yap1mScarlet , and examined YAP-mScarlet protein dynamics during the 8-32-cell stages using live imaging. During mitosis, YAP-mScarlet diffused throughout the cells. YAP-mScarlet dynamics in daughter cells varied depending on the cell division patterns. YAP-mScarlet localization in daughter cells at the completion of cell division coincided with that in mother cells. Experimental manipulation of YAP-mScarlet localization in mother cells also altered its localization in daughter cells upon completion of cell division. In daughter cells, YAP-mScarlet localization gradually changed to the final pattern. In some divisions during the 8-16-cell stages, the cytoplasmic YAP-mScarlet localization preceded cell internalization. These results suggest that cell position is not a primary determinant of YAP localization and that the Hippo signaling status of the mother cell is inherited by the daughter cells, which likely contributes to the stabilization of the cell fate specification process beyond cell division.
Collapse
Affiliation(s)
- Tomoaki Otsuka
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Hiromi Shimojo
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
35
|
Gahurova L, Tomankova J, Cerna P, Bora P, Kubickova M, Virnicchi G, Kovacovicova K, Potesil D, Hruska P, Zdrahal Z, Anger M, Susor A, Bruce AW. Spatial positioning of preimplantation mouse embryo cells is regulated by mTORC1 and m 7G-cap-dependent translation at the 8- to 16-cell transition. Open Biol 2023; 13:230081. [PMID: 37553074 PMCID: PMC10409569 DOI: 10.1098/rsob.230081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
Preimplantation mouse embryo development involves temporal-spatial specification and segregation of three blastocyst cell lineages: trophectoderm, primitive endoderm and epiblast. Spatial separation of the outer-trophectoderm lineage from the two other inner-cell-mass (ICM) lineages starts with the 8- to 16-cell transition and concludes at the 32-cell stages. Accordingly, the ICM is derived from primary and secondary contributed cells; with debated relative EPI versus PrE potencies. We report generation of primary but not secondary ICM populations is highly dependent on temporal activation of mammalian target of Rapamycin (mTOR) during 8-cell stage M-phase entry, mediated via regulation of the 7-methylguanosine-cap (m7G-cap)-binding initiation complex (EIF4F) and linked to translation of mRNAs containing 5' UTR terminal oligopyrimidine (TOP-) sequence motifs, as knockdown of identified TOP-like motif transcripts impairs generation of primary ICM founders. However, mTOR inhibition-induced ICM cell number deficits in early blastocysts can be compensated by the late blastocyst stage, after inhibitor withdrawal; compensation likely initiated at the 32-cell stage when supernumerary outer cells exhibit molecular characteristics of inner cells. These data identify a novel mechanism specifically governing initial spatial segregation of mouse embryo blastomeres, that is distinct from those directing subsequent inner cell formation, contributing to germane segregation of late blastocyst lineages.
Collapse
Affiliation(s)
- Lenka Gahurova
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
- Laboratory of Biochemistry and Molecular Biology of Germ Cells, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburská 89, 27721 Liběchov, Czech Republic
| | - Jana Tomankova
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Pavlina Cerna
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Pablo Bora
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Michaela Kubickova
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Giorgio Virnicchi
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Kristina Kovacovicova
- Laboratory of Cell Division Control, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburská 89, 27721 Liběchov, Czech Republic
- Department of Genetics and Reproduction, Central European Institute of Technology, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - David Potesil
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Pavel Hruska
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Zbynek Zdrahal
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Martin Anger
- Laboratory of Cell Division Control, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburská 89, 27721 Liběchov, Czech Republic
- Department of Genetics and Reproduction, Central European Institute of Technology, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - Andrej Susor
- Laboratory of Biochemistry and Molecular Biology of Germ Cells, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburská 89, 27721 Liběchov, Czech Republic
| | - Alexander W Bruce
- Laboratory of Early Mammalian Developmental Biology (LEMDB), Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| |
Collapse
|
36
|
Kim M, Lee J, Cai L, Choi H, Oh D, Jawad A, Hyun SH. Neurotrophin-4 promotes the specification of trophectoderm lineage after parthenogenetic activation and enhances porcine early embryonic development. Front Cell Dev Biol 2023; 11:1194596. [PMID: 37519302 PMCID: PMC10373506 DOI: 10.3389/fcell.2023.1194596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Neurotrophin-4 (NT-4), a neurotrophic factor, appears to affect early embryonic development because it is secreted not only by neurons but also by oviductal and uterine epithelial cells. However, no studies have characterized the effects of NT-4 on early embryonic development in pigs. In this study, we applied the experimental model of parthenogenetic-activation (PA)-derived embryos. Herein, we investigated the effect of NT-4 supplementation during the in vitro culture (IVC) of embryos, analyzed the transcription levels of specific genes, and outlined the first cell lineage specification for porcine PA-derived blastocysts. We confirmed that NT-4 and its receptor proteins were localized in both the inner cell mass (ICM) and trophectoderm (TE) in porcine blastocysts. Across different concentrations (0, 1, 10, and 100 ng/mL) of NT-4 supplementation, the optimal concentration of NT-4 to improve the developmental competence of porcine parthenotes was 10 ng/mL. NT-4 supplementation during porcine IVC significantly (p < 0.05) increased the proportion of TE cells by inducing the transcription of TE lineage markers (CDX2, PPAG3, and GATA3 transcripts). NT-4 also reduced blastocyst apoptosis by regulating the transcription of apoptosis-related genes (BAX and BCL2L1 transcripts) and improved blastocyst quality via the interaction of neurotrophin-, Hippo-yes-associated protein (Hippo-YAP) and mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK) pathway. Additionally, NT-4 supplementation during IVC significantly (p < 0.05) increased YAP1 transcript levels and significantly (p < 0.01) decreased LATS2 transcript levels, respectively, in the porcine PA-derived blastocysts. We also confirmed through fluorescence intensity that the YAP1 protein was significantly (p < 0.001) increased in the NT-4-treated blastocysts compared with that in the control. NT-4 also promoted differentiation into the TE lineage rather than into the ICM lineage during porcine early embryonic development. In conclusion, 10 ng/mL NT-4 supplementation enhanced blastocyst quality by regulating the apoptosis- and TE lineage specification-related genes and interacting with neurotrophin-, Hippo-YAP-, and MAPK/ERK signaling pathway during porcine in vitro embryo development.
Collapse
Affiliation(s)
- Mirae Kim
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Joohyeong Lee
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Lian Cai
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyerin Choi
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjin Oh
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Ali Jawad
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Hwan Hyun
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
37
|
Regin M, Essahib W, Demtschenko A, Dewandre D, David L, Gerri C, Niakan KK, Verheyen G, Tournaye H, Sterckx J, Sermon K, Van De Velde H. Lineage segregation in human pre-implantation embryos is specified by YAP1 and TEAD1. Hum Reprod 2023:7193343. [PMID: 37295962 DOI: 10.1093/humrep/dead107] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/02/2023] [Indexed: 06/12/2023] Open
Abstract
STUDY QUESTION Which processes and transcription factors specify the first and second lineage segregation events during human preimplantation development? SUMMARY ANSWER Differentiation into trophectoderm (TE) cells can be initiated independently of polarity; moreover, TEAD1 and YAP1 co-localize in (precursor) TE and primitive endoderm (PrE) cells, suggesting a role in both the first and the second lineage segregation events. WHAT IS KNOWN ALREADY We know that polarity, YAP1/GATA3 signalling and phospholipase C signalling play a key role in TE initiation in compacted human embryos, however, little is known about the TEAD family of transcription factors that become activated by YAP1 and, especially, whether they play a role during epiblast (EPI) and PrE formation. In mouse embryos, polarized outer cells show nuclear TEAD4/YAP1 activity that upregulates Cdx2 and Gata3 expression while inner cells exclude YAP1 which upregulates Sox2 expression. The second lineage segregation event in mouse embryos is orchestrated by FGF4/FGFR2 signalling which could not be confirmed in human embryos; TEAD1/YAP1 signalling also plays a role during the establishment of mouse EPI cells. STUDY DESIGN, SIZE, DURATION Based on morphology, we set up a development timeline of 188 human preimplantation embryos between Day 4 and 6 post-fertilization (dpf). The compaction process was divided into three subgroups: embryos at the start (C0), during (C1), and at the end (C2) of, compaction. Inner cells were identified as cells that were entirely separated from the perivitelline space and enclosed by cellular contacts on all sides. The blastulation process was divided into six subgroups, starting with early blastocysts with sickle-cell shaped outer cells (B0) and further on, blastocysts with a cavity (B1). Full blastocysts (B2) showed a visible ICM and outer cells referred to as TE. Further expanded blastocysts (B3) had accumulated fluid and started to expand due to TE cell proliferation and zona pellucida (ZP) thinning. The blastocysts then significantly expanded further (B4) and started to hatch out of the ZP (B5) until they were fully hatched (B6). PARTICIPANTS/MATERIALS, SETTING, METHODS After informed consent and the expiration of the 5-year cryopreservation duration, 188 vitrified high quality eight-cell stage human embryos (3 dpf) were warmed and cultured until the required stages were reached. We also cultured 14 embryos that were created for research until the four- and eight-cell stage. The embryos were scored according to their developmental stage (C0-B6) displaying morphological key differences, rather than defining them according to their chronological age. They were fixed and immunostained for different combinations of cytoskeleton (F-actin), polarization (p-ERM), TE (GATA3), EPI (NANOG), PrE (GATA4 and SOX17), and members of the Hippo signalling pathway (YAP1, TEAD1 and TEAD4). We choose these markers based on previous observations in mouse embryos and single cell RNA-sequencing data of human embryos. After confocal imaging (LSM800, Zeiss), we analysed cell numbers within each lineage, different co-localization patterns and nuclear enrichment. MAIN RESULTS AND THE ROLE OF CHANCE We found that in human preimplantation embryos compaction is a heterogeneous process that takes place between the eight-cell to the 16-cell stages. Inner and outer cells are established at the end of the compaction process (C2) when the embryos contain up to six inner cells. Full apical p-ERM polarity is present in all outer cells of compacted C2 embryos. Co-localization of p-ERM and F-actin increases steadily from 42.2% to 100% of the outer cells, between C2 and B1 stages, while p-ERM polarizes before F-actin (P < 0.00001). Next, we sought to determine which factors specify the first lineage segregation event. We found that 19.5% of the nuclei stain positive for YAP1 at the start of compaction (C0) which increases to 56.1% during compaction (C1). At the C2 stage, 84.6% of polarized outer cells display high levels of nuclear YAP1 while it is absent in 75% of non-polarized inner cells. In general, throughout the B0-B3 blastocyst stages, polarized outer/TE cells are mainly positive for YAP1 and non-polarized inner/ICM cells are negative for YAP1. From the C1 stage onwards, before polarity is established, the TE marker GATA3 is detectable in YAP1 positive cells (11.6%), indicating that differentiation into TE cells can be initiated independently of polarity. Co-localization of YAP1 and GATA3 increases steadily in outer/TE cells (21.8% in C2 up to 97.3% in B3). Transcription factor TEAD4 is ubiquitously present throughout preimplantation development from the compacted stage onwards (C2-B6). TEAD1 displays a distinct pattern that coincides with YAP1/GATA3 co-localization in the outer cells. Most outer/TE cells throughout the B0-B3 blastocyst stages are positive for TEAD1 and YAP1. However, TEAD1 proteins are also detected in most nuclei of the inner/ICM cells of the blastocysts from cavitation onwards, but at visibly lower levels as compared to that in TE cells. In the ICM of B3 blastocysts, we found one main population of cells with NANOG+/SOX17-/GATA4- nuclei (89.1%), but exceptionally we found NANOG+/SOX17+/GATA4+ cells (0.8%). In seven out of nine B3 blastocysts, nuclear NANOG was found in all the ICM cells, supporting the previously reported hypothesis that PrE cells arise from EPI cells. Finally, to determine which factors specify the second lineage segregation event, we co-stained for TEAD1, YAP1, and GATA4. We identified two main ICM cell populations in B4-6 blastocysts: the EPI (negative for the three markers, 46.5%) and the PrE (positive for the three markers, 28.1%) cells. We conclude that TEAD1 and YAP1 co-localise in (precursor) TE and PrE cells, indicating that TEAD1/YAP1 signalling plays a role in the first and the second lineage segregation events. LIMITATIONS, REASONS FOR CAUTION In this descriptive study, we did not perform functional studies to investigate the role of TEAD1/YAP1 signalling during the first and second lineage segregation events. WIDER IMPLICATIONS OF THE FINDINGS Our detailed roadmap on polarization, compaction, position and lineage segregation events during human preimplantation development paves the way for further functional studies. Understanding the gene regulatory networks and signalling pathways involved in early embryogenesis could ultimately provide insights into why embryonic development is sometimes impaired and facilitate the establishment of guidelines for good practice in the IVF lab. STUDY FUNDING/COMPETING INTERESTS This work was financially supported by Wetenschappelijk Fonds Willy Gepts (WFWG) of the University Hospital UZ Brussel (WFWG142) and the Fonds Wetenschappelijk Onderzoek-Vlaanderen (FWO, G034514N). M.R. is doctoral fellow at the FWO. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Marius Regin
- Research Group Reproduction and Genetics (REGE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Wafaa Essahib
- Research Group Reproduction and Immunology (REIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Andrej Demtschenko
- Research Group Reproduction and Genetics (REGE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Delphine Dewandre
- Research Group Reproduction and Genetics (REGE), Vrije Universiteit Brussel, Brussels, Belgium
- Beacon CARE Fertility, Beacon Consultants Concourse, Sandyford, Dublin, Ireland
| | - Laurent David
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
- Université de Nantes, CHU Nantes, INSERM, CNRS, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Claudia Gerri
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, London, UK
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, Dresden, 01307, Germany
| | - Kathy K Niakan
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, London, UK
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, Cambridge, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Epigenetics Programme, Babraham Institute, Cambridge, UK
| | - Greta Verheyen
- Brussels IVF, Universitair Ziekenhuis Brussel, Belgium, Brussels
| | - Herman Tournaye
- Brussels IVF, Universitair Ziekenhuis Brussel, Belgium, Brussels
- Department of Obstetrics, Gynaecology, Perinatology and Reproduction, Institute of Professional Education, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Johan Sterckx
- Brussels IVF, Universitair Ziekenhuis Brussel, Belgium, Brussels
| | - Karen Sermon
- Research Group Reproduction and Genetics (REGE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Hilde Van De Velde
- Research Group Reproduction and Immunology (REIM), Vrije Universiteit Brussel, Brussels, Belgium
- Brussels IVF, Universitair Ziekenhuis Brussel, Belgium, Brussels
| |
Collapse
|
38
|
Lackner A, Müller M, Gamperl M, Stoeva D, Langmann O, Papuchova H, Roitinger E, Dürnberger G, Imre R, Mechtler K, Latos PA. The Fgf/Erf/NCoR1/2 repressive axis controls trophoblast cell fate. Nat Commun 2023; 14:2559. [PMID: 37137875 DOI: 10.1038/s41467-023-38101-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/15/2023] [Indexed: 05/05/2023] Open
Abstract
Placental development relies on coordinated cell fate decisions governed by signalling inputs. However, little is known about how signalling cues are transformed into repressive mechanisms triggering lineage-specific transcriptional signatures. Here, we demonstrate that upon inhibition of the Fgf/Erk pathway in mouse trophoblast stem cells (TSCs), the Ets2 repressor factor (Erf) interacts with the Nuclear Receptor Co-Repressor Complex 1 and 2 (NCoR1/2) and recruits it to key trophoblast genes. Genetic ablation of Erf or Tbl1x (a component of the NCoR1/2 complex) abrogates the Erf/NCoR1/2 interaction. This leads to mis-expression of Erf/NCoR1/2 target genes, resulting in a TSC differentiation defect. Mechanistically, Erf regulates expression of these genes by recruiting the NCoR1/2 complex and decommissioning their H3K27ac-dependent enhancers. Our findings uncover how the Fgf/Erf/NCoR1/2 repressive axis governs cell fate and placental development, providing a paradigm for Fgf-mediated transcriptional control.
Collapse
Affiliation(s)
- Andreas Lackner
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Michael Müller
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Magdalena Gamperl
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Delyana Stoeva
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Olivia Langmann
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Henrieta Papuchova
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | | | | | - Richard Imre
- Institute of Molecular Pathology, A-1030, Vienna, Austria
| | - Karl Mechtler
- Institute of Molecular Pathology, A-1030, Vienna, Austria
| | - Paulina A Latos
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria.
| |
Collapse
|
39
|
Chen ACH, Lee YL, Ruan H, Huang W, Fong SW, Tian S, Lee KC, Wu GM, Tan Y, Wong TCH, Wu J, Zhang W, Cao D, Chow JFC, Liu P, Yeung WSB. Expanded Potential Stem Cells from Human Embryos Have an Open Chromatin Configuration with Enhanced Trophoblast Differentiation Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204797. [PMID: 36775869 PMCID: PMC10104645 DOI: 10.1002/advs.202204797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Human expanded potential stem cells (hEPSC) have been derived from human embryonic stem cells and induced pluripotent stem cells. Here direct derivation of hEPSC from human pre-implantation embryos is reported. Like the reported hEPSC, the embryo-derived hEPSC (hEPSC-em) exhibit a transcriptome similar to morula, comparable differentiation potency, and high genome editing efficiency. Interestingly, the hEPSC-em show a unique H3 lysine-4 trimethylation (H3K4me3) open chromatin conformation; they possess a higher proportion of H3K4me3 bound broad domain (>5 kb) than the reported hEPSC, naive, and primed embryonic stem cells. The open conformation is associated with enhanced trophoblast differentiation potency with increased trophoblast gene expression upon induction of differentiation and success in derivation of trophoblast stem cells with bona fide characteristics. Hippo signaling is specifically enriched in the H3K4me3 broad domains of the hEPSC-. Knockout of the Hippo signaling gene, YAP1 abolishes the ability of the embryo-derived EPSC to form trophoblast stem cells.
Collapse
Affiliation(s)
- Andy Chun Hang Chen
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Hanzhang Ruan
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Wen Huang
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Siyu Tian
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Kai Chuen Lee
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Genie Minju Wu
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Yongqi Tan
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Timothy Chun Hin Wong
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Jian Wu
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Weiyu Zhang
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Dandan Cao
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
| | - Judy Fung Cheung Chow
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Pengtao Liu
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongStem Cell and Regenerative Medicine ConsortiumHong KongHong Kong
| | - William Shu Biu Yeung
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| |
Collapse
|
40
|
The transcription factor ELF5 is essential for early preimplantation development. Mol Biol Rep 2023; 50:2119-2125. [PMID: 36542237 DOI: 10.1007/s11033-022-08217-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND During early embryonic development, the cell adhesion molecule E-cadherin encoded by the Cdh1 gene plays a vital role in providing proper cell-cell adhesion, ensuring an undifferentiated state critical for maintaining the pluripotency for the development of the preimplantation embryo. The transcriptional regulation of Cdh1 gained attention recently but is not yet fully understood. In a previous study, our team established a correlation between Elf3 and Cdh1 expression and showed its importance in the regulation of MET. METHODS AND RESULTS Here, the regulation of Cdh1 by Ets transcription factors in early embryogenesis was investigated. A loss-of-function approach was used to study the effect of Elf5 loss on Cdh1 gene expression by small interfering RNAs in fertilized oocytes. Changes in gene expression were measured by qPCR analysis, and developing embryos were visualized by microscopy. Loss of Elf5 arrested the embryos at the 2-cell stage, accompanied by a significant downregulation of Cdh1 expression. CONCLUSION The findings presented here illustrate the role of ELF5 in preimplantation development and in regulating the expression of Cdh1. The maintenance of the ELF5 and Cdh1 regulatory node proved essential for the proper development of the early mouse embryos, which is in agreement with the critical role of Elf5 and Cdh1 genes in regulating the early events during embryogenesis.
Collapse
|
41
|
Cheng Y, Xiao Y, Ruan Y, Wang J, Tian Y, Xiong J, Wang J, Wang F, Zhang C, Xu Y, Liu L, Yu M, Wang J, Zhao B, Zhang Y, Yang R, Yang Y, Yao Z, Jian R, Xiao L, Zhang J. Comparative expression analysis of TEADs and their splice variants in mouse embryonic stem cells. Gene Expr Patterns 2023; 47:119302. [PMID: 36516960 DOI: 10.1016/j.gep.2022.119302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Transcriptional enhanced associate domain (TEAD) transcription factors play important roles in embryonic stem cell (ESC) renewal and differentiation. Four TEAD transcription factors (Tead1, Tead2, Tead3 and Tead4) and their various splice variants have been discovered in mice, but the expression pattern of them during pluripotency state transition is unclear. Here, we investigated the expression of TEADs and their splice variants in mouse ESCs at different pluripotent/differentiating states and adult mouse tissues. Our results preliminarily revealed the diversity and heterogeneity of TEAD family, which is helpful for understanding their overlapping and distinctive functions. Furthermore, a novel splice variant of Tead1 was identified and named Tead1 isoform 4.
Collapse
Affiliation(s)
- Yuda Cheng
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Yang Xiao
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Yan Ruan
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Jiali Wang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Yanping Tian
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Jiaxiang Xiong
- Experimental Center of Basic Medicine, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Jiaqi Wang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Institute of Immunology PLA & Department of Immunology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Fengsheng Wang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Chen Zhang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Yixiao Xu
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Southwest Eye Hospital, Southwest Hospital, the First Hospital Affiliated to Army Medical University, Chongqing, 400038, China
| | - Lianlian Liu
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Meng Yu
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Department of Joint Surgery, Southwest Hospital, the First Hospital Affiliated to Army Medical University, Chongqing, 400038, China
| | - Jiangjun Wang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Department of Cell Biology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Binyu Zhao
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Department of Physiology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Yue Zhang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Southwest Eye Hospital, Southwest Hospital, the First Hospital Affiliated to Army Medical University, Chongqing, 400038, China
| | - Ran Yang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Department of Pathophysiology, College of High-Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Zhongxiang Yao
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China; Department of Physiology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Rui Jian
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China.
| | - Junlei Zhang
- Laboratory of Stem Cell and Developmental Biology, Department of Histology and Embryology, College of Basic Medical Science, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
42
|
Machtinger R, Racowsky C, Baccarelli AA, Bollati V, Orvieto R, Hauser R, Barnett-Itzhaki Z. Recombinant human chorionic gonadotropin and gonadotropin-releasing hormone agonist differently affect the profile of extracellular vesicle microRNAs in human follicular fluid. J Assist Reprod Genet 2023; 40:527-536. [PMID: 36609942 PMCID: PMC10033801 DOI: 10.1007/s10815-022-02703-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 12/19/2022] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To compare the expression profile of extracellular vesicle microRNAs (EV-miRNAs) derived from follicular fluid after a trigger with recombinant human chorionic gonadotropin (r-hCG) or with a gonadotropin-releasing hormone GnRH agonist (GnRH-a) for final oocyte maturation. METHODS A retrospective analysis of a prospective cohort. Women undergoing in vitro fertilization at a tertiary university-affiliated hospital were recruited between 2014 and 2016. EV-miRNAs were extracted from the follicular fluid of a single follicle, and their expression was assessed using TaqMan Open Array®. Genes regulated by EV-miRNAs were analyzed using miRWalk2.0 Targetscan database, DAVID Bioinformatics Resources, Kyoto-Encyclopedia of Genes and Genomes (KEGG), and Gene Ontology (GO). RESULTS Eighty-two women were included in the r-hCG trigger group and 9 in the GnRH-a group. Of 754 EV-miRNAs screened, 135 were detected in at least 50% of the samples and expressed in both groups and were further analyzed. After adjusting for multiple testing, 41 EV-miRNAs whose expression levels significantly differed between the two trigger groups were identified. Bioinformatics analysis of the genes regulated by these EV-miRNAs showed distinct pathways between the two triggers, including TGF-beta signaling, cell cycle, and Wnt signaling pathways. Most of these pathways regulate cascades associated with apoptosis, embryo development, implantation, decidualization, and placental development. CONCLUSIONS Trigger with GnRH-a or r-hCG leads to distinct EV-miRNAs expression profiles and to downstream biological effects in ovarian follicles. These findings may provide an insight for the increased apoptosis and the lower implantation rates following GnRH-a trigger vs. r-hCG in cases lacking intensive luteal phase support.
Collapse
Affiliation(s)
- R Machtinger
- Department of Obstetrics and Gynecology, Division of IVF, Sheba Medical Center, Ramat Gan 5262000, Israel.
- Sackler School of Medicine, Tel-Aviv University, 6997801, Tel Aviv, Israel.
| | - C Racowsky
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hospital Foch, Suresnes, France
| | - A A Baccarelli
- Laboratory of Precision Environmental Biosciences, Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY, 10032, USA
| | - V Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, 20122, Milan, Italy
- Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - R Orvieto
- Department of Obstetrics and Gynecology, Division of IVF, Sheba Medical Center, Ramat Gan 5262000, Israel
| | - R Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Z Barnett-Itzhaki
- Public Health Services, Ministry of Health, 9446724, Jerusalem, Israel
- Faculty of Engineering, Ruppin Academic Center, 4025000, Emek Hefer, Israel
- Ruppin Research Group in Environmental and Social Sustainability, Ruppin Academic Center, 4025000, Emek Hefer, Israel
| |
Collapse
|
43
|
Zhuge R, Wang C, Wang J, Yu S, Liao L, Zheng X. hCINAP regulates the differentiation of embryonic stem cells by regulating NEDD4 liquid-liquid phase-separation-mediated YAP1 activation. Cell Rep 2023; 42:111935. [PMID: 36640330 DOI: 10.1016/j.celrep.2022.111935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
YAP1 functions in lineage differentiation of pluripotent embryonic stem cells (ESCs); however, the detailed mechanisms underlying the regulation of YAP1 activity during ESC differentiation remain elusive. Here, we report that hCINAP serves as a negative regulator of YAP1 during ESC fate decisions. The expression of mCINAP, the murine homolog of hCINAP, is downregulated during the differentiation process of murine ESC (mESC) ectoderm lineage, leading to liquid-liquid phase separation (LLPS) of NEDD4 and activation of YAP1. Mechanistically, hCINAP interacts with and prevents NEDD4 from forming cytoplasmic condensates that compartmentalize YAP1 and its kinase NLK, facilitating YAP1 phosphorylation at Ser128 and promoting YAP1 activation. mCINAP depletion leads to the formation of NEDD4 condensates and YAP1 activation, which impedes endoderm differentiation of mESCs. Our study shows that hCINAP is a vital regulator of YAP1 activity and is essential for stem cell fate decisions, which provides mechanistic insight into early embryogenesis.
Collapse
Affiliation(s)
- Ruipeng Zhuge
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Chao Wang
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Jie Wang
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Shuyu Yu
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Liming Liao
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiaofeng Zheng
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing 100871, China; Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
44
|
Lee SJ, Kim J, Han G, Hong SP, Kim D, Cho C. Impaired Blastocyst Formation in Lnx2-Knockdown Mouse Embryos. Int J Mol Sci 2023; 24:ijms24021385. [PMID: 36674899 PMCID: PMC9867088 DOI: 10.3390/ijms24021385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Ligand of Numb-protein X 2 (LNX2) is an E3 ubiquitin ligase that is known to regulate Notch signaling by participating in NUMB protein degradation. Notch signaling is important for differentiation and proliferation in mammals, and plays a significant role in blastocyst formation during early embryonic development. In this study, we investigated Lnx2 in mouse preimplantation embryos. Expression analysis showed that Lnx2 is expressed in oocytes and preimplantation embryos. Lnx2-knockdown embryos normally progress to the morula stage, but the majority of them do not develop into normal blastocysts. Transcript analysis revealed that the expression levels of genes critical for cell lineage specification, including octamer-binding transcription factor 4 (Oct4), are increased in Lnx2 knockdown embryos. Furthermore, the expression levels of Notch and Hippo signaling-related genes are also increased by Lnx2 knockdown. Collectively, our results show that Lnx2 is important for blastocyst formation in mice, suggest that this may act via lineage specification of inner cell mass, and further show that Lnx2 may be involved in transcriptionally regulating various genes implicated in early embryonic development.
Collapse
Affiliation(s)
- Seung-Jae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jaehwan Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Developmental Epigenetics Laboratory, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Gwidong Han
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Seung-Pyo Hong
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dayeon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Correspondence:
| |
Collapse
|
45
|
Goissis MD, Cibelli JB. Early Cell Specification in Mammalian Fertilized and Somatic Cell Nuclear Transfer Embryos. Methods Mol Biol 2023; 2647:59-81. [PMID: 37041329 DOI: 10.1007/978-1-0716-3064-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Early cell specification in mammalian preimplantation embryos is an intricate cellular process that leads to coordinated spatial and temporal expression of specific genes. Proper segregation into the first two cell lineages, the inner cell mass (ICM) and the trophectoderm (TE), is imperative for developing the embryo proper and the placenta, respectively. Somatic cell nuclear transfer (SCNT) allows the formation of a blastocyst containing both ICM and TE from a differentiated cell nucleus, which means that this differentiated genome must be reprogrammed to a totipotent state. Although blastocysts can be generated efficiently through SCNT, the full-term development of SCNT embryos is impaired mostly due to placental defects. In this review, we examine the early cell fate decisions in fertilized embryos and compare them to observations in SCNT-derived embryos, in order to understand if these processes are affected by SCNT and could be responsible for the low success of reproductive cloning.
Collapse
Affiliation(s)
- Marcelo D Goissis
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, SP, Brazil.
| | - Jose B Cibelli
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
46
|
Lamba A, Zernicka-Goetz M. The role of polarization and early heterogeneities in the mammalian first cell fate decision. Curr Top Dev Biol 2023; 154:169-196. [PMID: 37100517 DOI: 10.1016/bs.ctdb.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
The first cell fate decision is the process by which cells of an embryo take on distinct lineage identities for the first time, representing the beginning of patterning during development. In mammals, this process separates an embryonic inner cell mass lineage (future new organism) from an extra-embryonic trophectoderm lineage (future placenta), and in the mouse, this is classically attributed to the consequences of apical-basal polarity. The mouse embryo acquires this polarity at the 8-cell stage, indicated by cap-like protein domains on the apical surface of each cell; those cells which retain polarity over subsequent divisions are specified as trophectoderm, and the rest as inner cell mass. Recent research has advanced our knowledge of this process - this review will discuss mechanisms behind the establishment of polarity and distribution of the apical domain, different factors affecting the first cell fate decision including heterogeneities between cells of the very early embryo, and the conservation of developmental mechanisms across species, including human.
Collapse
Affiliation(s)
- Adiyant Lamba
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
47
|
Liu W, Chen J, Yang C, Lee KF, Lee YL, Chiu PCN, Zhang Y, Duan YG, Liu K, Yeung WSB. Expression of microRNA let-7 in cleavage embryos modulates cell fate determination and formation of mouse blastocysts†. Biol Reprod 2022; 107:1452-1463. [PMID: 36136742 DOI: 10.1093/biolre/ioac181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/30/2022] [Accepted: 09/14/2022] [Indexed: 12/24/2022] Open
Abstract
After fertilization, the zygote undergoes cell division. Up to the 8-cell stage, the blastomeres of mouse preimplantation embryos are morphologically identical. The first cell differentiation starts in the morula leading to the formation of trophectoderm cells and inner cell mass cells of the blastocyst. The regulation of the differentiation event and the formation of blastocysts are not fully known. Lethal-7 (let-7) is a family of evolutionarily conserved microRNAs. Here, we showed that the expression of let-7a and let-7g decreased drastically from the 1-cell stage to the 2-cell stage, remained low up to the 8-cell stage and slightly increased after the morula stage of mouse embryos. The expression of let-7 in the inner cell mass was higher than that in the trophectoderm. Forced expression of let-7a in embryos at the 1-cell and 4-cell stage inhibited blastocyst formation and downregulated the expression of CDX2 but maintained that of OCT4 in the trophectoderm. Forced expression of other let-7 isoforms exhibited similar inhibitory action on blastulation. On the other hand, inhibition of let-7a at the 4-cell stage and the 8-cell stage enhanced blastocyst formation. Co-injection of green fluorescent protein (GFP) mRNA (lineage tracer) with either precursor of let-7a (pre-let-7a) or scramble control into one blastomere of 2-cell embryos showed that ~75% of the resulting blastocysts possessed GFP+ cells in their inner cell mass only. The biased development towards the inner cell mass with forced expression of let-7 was reproduced in 2-cell chimeric embryos consisting of one wildtype blastomere and one GFP mRNA-injected blastomere from another 2-cell embryo carrying a doxycycline-inducible let-7g gene. Bioinformatics analysis indicated that Tead4 was a potential target of let-7. Let-7 bound to the 3'UTR of Tead4 and let-7 forced expression downregulated the expression of Tead4 in mouse blastocysts. Co-injection of Tead4 mRNA partially nullified the modulatory roles of let-7a in the inner cell mass cell fate. In conclusion, a high level of let-7 at the 2-cell stage favored the formation of the inner cell mass, whereas a low level of let-7 at the 4-cell to 8-cell stage enhanced blastocyst formation. Tead4 mediated the action of let-7 on the inner cell mass cell-fate determination.
Collapse
Affiliation(s)
- Weimin Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jing Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Yang
- Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yin-Lau Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Philip Chi-Ngong Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yihui Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gong Duan
- Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kui Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - William Shu-Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Center of Reproductive Medicine and Prenatal Diagnosis, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
48
|
Luo M, Xu Y, Chen H, Wu Y, Pang A, Hu J, Dong X, Che J, Yang H. Advances of targeting the YAP/TAZ-TEAD complex in the hippo pathway for the treatment of cancers. Eur J Med Chem 2022; 244:114847. [DOI: 10.1016/j.ejmech.2022.114847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
49
|
Hazra R, Brine L, Garcia L, Benz B, Chirathivat N, Shen MM, Wilkinson JE, Lyons SK, Spector DL. Platr4 is an early embryonic lncRNA that exerts its function downstream on cardiogenic mesodermal lineage commitment. Dev Cell 2022; 57:2450-2468.e7. [PMID: 36347239 PMCID: PMC9680017 DOI: 10.1016/j.devcel.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/22/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
The mammalian genome encodes thousands of long non-coding RNAs (lncRNAs), many of which are developmentally regulated and differentially expressed across tissues, suggesting their potential roles in cellular differentiation. Despite this expression pattern, little is known about how lncRNAs influence lineage commitment at the molecular level. Here, we demonstrate that perturbation of an embryonic stem cell/early embryonic lncRNA, pluripotency-associated transcript 4 (Platr4), directly influences the specification of cardiac-mesoderm-lineage differentiation. We show that Platr4 acts as a molecular scaffold or chaperone interacting with the Hippo-signaling pathway molecules Yap and Tead4 to regulate the expression of a downstream target gene, Ctgf, which is crucial to the cardiac-lineage program. Importantly, Platr4 knockout mice exhibit myocardial atrophy and valve mucinous degeneration, which are both associated with reduced cardiac output and sudden heart failure. Together, our findings provide evidence that Platr4 is required in cardiac-lineage specification and adult heart function in mice.
Collapse
Affiliation(s)
- Rasmani Hazra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lily Brine
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Libia Garcia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Brian Benz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Napon Chirathivat
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael M Shen
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Scott K Lyons
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David L Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
50
|
Tomikawa J, Miyamoto K. Structural alteration of the nucleus for the reprogramming of gene expression. FEBS J 2022; 289:7221-7233. [PMID: 33891358 DOI: 10.1111/febs.15894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/06/2021] [Accepted: 04/19/2021] [Indexed: 01/13/2023]
Abstract
The regulation of gene expression is a critical process for establishing and maintaining cellular identity. Gene expression is controlled through a chromatin-based mechanism in the nucleus of eukaryotic cells. Recent studies suggest that chromatin accessibility and the higher-order structure of chromatin affect transcriptional outcome. This is especially evident when cells change their fate during development and nuclear reprogramming. Furthermore, non-chromosomal contents of the cell nucleus, namely nucleoskeleton proteins, can also affect chromatin and nuclear structures, resulting in transcriptional alterations. Here, we review our current mechanistic understanding about how chromatin and nuclear structures impact transcription in the course of embryonic development, cellular differentiation and nuclear reprogramming, and also discuss unresolved questions that remain to be addressed in the field.
Collapse
Affiliation(s)
- Junko Tomikawa
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| | - Kei Miyamoto
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| |
Collapse
|