1
|
Shukla V, Moreno-Irusta A, Varberg KM, Kuna M, Iqbal K, Galligos AM, Aplin JD, Choudhury RH, Okae H, Arima T, Soares MJ. NOTUM-mediated WNT silencing drives extravillous trophoblast cell lineage development. Proc Natl Acad Sci U S A 2024; 121:e2403003121. [PMID: 39325428 PMCID: PMC11459147 DOI: 10.1073/pnas.2403003121] [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: 02/14/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Trophoblast stem (TS) cells have the unique capacity to differentiate into specialized cell types, including extravillous trophoblast (EVT) cells. EVT cells invade into and transform the uterus where they act to remodel the vasculature facilitating the redirection of maternal nutrients to the developing fetus. Disruptions in EVT cell development and function are at the core of pregnancy-related disease. WNT-activated signal transduction is a conserved regulator of morphogenesis of many organ systems, including the placenta. In human TS cells, activation of canonical WNT signaling is critical for maintenance of the TS cell stem state and its downregulation accompanies EVT cell differentiation. We show that aberrant WNT signaling undermines EVT cell differentiation. Notum, palmitoleoyl-protein carboxylesterase (NOTUM), a negative regulator of canonical WNT signaling, was prominently expressed in first-trimester EVT cells developing in situ and up-regulated in EVT cells derived from human TS cells. Furthermore, NOTUM was required for optimal human TS cell differentiation to EVT cells. Activation of NOTUM in EVT cells is driven, at least in part, by endothelial Per-Arnt-Sim (PAS) domain 1 (also called hypoxia-inducible factor 2 alpha). Collectively, our findings indicate that canonical Wingless-related integration site (WNT) signaling is essential for maintenance of human trophoblast cell stemness and regulation of human TS cell differentiation. Downregulation of canonical WNT signaling via the actions of NOTUM is required for optimal EVT cell differentiation.
Collapse
Affiliation(s)
- Vinay Shukla
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Ayelen Moreno-Irusta
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Kaela M. Varberg
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Marija Kuna
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Khursheed Iqbal
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Anna M. Galligos
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - John D. Aplin
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, The University of Manchester, ManchesterM13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St. Mary’s Hospital, University of Manchester, ManchesterM13 9WL, United Kingdom
| | - Ruhul H. Choudhury
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, The University of Manchester, ManchesterM13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St. Mary’s Hospital, University of Manchester, ManchesterM13 9WL, United Kingdom
| | - Hiroaki Okae
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Michael J. Soares
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, MO64108
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
2
|
Shukla V, Moreno-Irusta A, Varberg KM, Kuna M, Iqbal K, Galligos AM, Aplin JD, Choudhury RH, Okae H, Arima T, Soares MJ. NOTUM-MEDIATED WNT SILENCING DRIVES EXTRAVILLOUS TROPHOBLAST CELL LINEAGE DEVELOPMENT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.579974. [PMID: 38405745 PMCID: PMC10888853 DOI: 10.1101/2024.02.13.579974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Trophoblast stem (TS) cells have the unique capacity to differentiate into specialized cell types, including extravillous trophoblast (EVT) cells. EVT cells invade into and transform the uterus where they act to remodel the vasculature facilitating the redirection of maternal nutrients to the developing fetus. Disruptions in EVT cell development and function are at the core of pregnancy-related disease. WNT-activated signal transduction is a conserved regulator of morphogenesis of many organ systems, including the placenta. In human TS cells, activation of canonical WNT signaling is critical for maintenance of the TS cell stem state and its downregulation accompanies EVT cell differentiation. We show that aberrant WNT signaling undermines EVT cell differentiation. Notum, palmitoleoyl-protein carboxylesterase (NOTUM), a negative regulator of canonical WNT signaling, was prominently expressed in first trimester EVT cells developing in situ and upregulated in EVT cells derived from human TS cells. Furthermore, NOTUM was required for optimal human TS cell differentiation to EVT cells. Activation of NOTUM in EVT cells is driven, at least in part, by endothelial PAS domain 1 (also called hypoxia-inducible factor 2 alpha). Collectively, our findings indicate that canonical WNT signaling is essential for maintenance of human trophoblast cell stemness and regulation of human TS cell differentiation. Downregulation of canonical WNT signaling via the actions of NOTUM is required for optimal EVT cell differentiation.
Collapse
Affiliation(s)
- Vinay Shukla
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Ayelen Moreno-Irusta
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Kaela M. Varberg
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Marija Kuna
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Khursheed Iqbal
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Anna M. Galligos
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John D. Aplin
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, The University of Manchester, Manchester M13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St Mary’s Hospital, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Ruhul H. Choudhury
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, The University of Manchester, Manchester M13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St Mary’s Hospital, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Hiroaki Okae
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811 Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, MO
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
3
|
Jiang LL, Yang DL, Han Q, Zhang HL, Pan M, Yan JY. LncRNA-NEAT1 blocks the Wnt/β-catenin signaling pathway by targeting miR-217 to inhibit trophoblast cell migration and invasion. J Assist Reprod Genet 2024; 41:2107-2115. [PMID: 38709402 PMCID: PMC11338999 DOI: 10.1007/s10815-024-03124-7] [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: 11/14/2023] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE This study aimed to study the correlation between preeclampsia (PE) and lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1), and to examine the molecular mechanisms behind the development of PE. METHODS 30 PE and 30 normal pregnant women placental samples were assessed the levels of NEAT1 and miR-217 by quantitative real-time PCR (qRT-PCR). The trophoblast cell line HTR8/SVneo was used for silencing NEAT1 or miR-217 inhibitor in the absence or presence of an inhibitor and H2O2. Cell counting Kit 8 (CCK-8), flow cytometry, and Transwell were used to detect cell proliferation, apoptosis, migration, and invasion. Luciferase reporter gene assay was utilized to verify the binding between miR-217 and Wnt family member 3 (Wnt3), and between the miR-217 and NEAT1. Proteins related to the Wnt/β-catenin signaling pathway were detected using western blotting. RESULTS The PE group exhibited a significantly downregulated expression of miR-217 and a significantly upregulated expression of NEAT1. NEAT1 targeted miR-217, and Wnt is a miR-217 target gene. siRNA-NEAT1 inhibited the apoptosis of trophoblast cells, but promoted their invasion, migration, and proliferation. MiR-217 inhibitor could partially reverse the effects of siRNA-NEAT1. The expression of the Wnt/β-catenin signaling pathway-related proteins, WNT signaling pathway inhibitor 1 (DKK1), cyclin-D1 and β-catenin, was significantly increased after siRNA-NEAT1. CONCLUSIONS NEAT1 could reduce trophoblast cell invasion and migration by suppressing miR-217/Wnt signaling pathway, leading to PE.
Collapse
Affiliation(s)
- Ling-Ling Jiang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Dan-Lin Yang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Qing Han
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Hua-le Zhang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Mian Pan
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China.
| | - Jian-Ying Yan
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China.
| |
Collapse
|
4
|
Xia S, Yu D, Wang Y, He B, Rong Y, Chen S, Xiao Z, Wang H, Wu H, Yan L. ISRIB facilitates the co-culture of human trophoblast stem cells and embryonic stem cells. Cell Prolif 2024; 57:e13599. [PMID: 38217296 DOI: 10.1111/cpr.13599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024] Open
Abstract
The embryo-like structures (embryoids) constructed by aggregating embryonic stem cells (ESCs) and trophoblast stem cells (TSCs) have provided revolutionary tools for studying the intricate interaction between embryonic and extra-embryonic tissues during early embryonic development, which has been achieved in mice. However, due to the opposite dependence on some signalling pathways for in vitro culture of human ESCs (hESCs) and TSCs (hTSCs), particularly WNT and TGFβ signalling pathways, which limits the construction of human post-implantation embryoids by aggregating hESCs and hTSCs. To overcome this challenge, here, by screening 1639 chemicals, we found that an inhibitor of integrated stress response, ISRIB, can replace WNT agonists and TGFβ inhibitors to maintain the stemness and differentiation capacity of hTSCs. Thus, we developed an ISRIB-dependent in vitro culture medium for hTSCs, namely nTSM. Furthermore, we demonstrated that ISRIB could also maintain the hESC stemness. Using a 3D co-culture system (hESCs and hTSCs aggregate, ETA), we demonstrated that a 1:1 mixture of hESC culture medium (ESM) and nTSM improved the cell proliferation and organisation of both hESC- and hTSC-compartments and the lumenogenesis of hESC-compartment in ETAs. Overall, our study provided an ISRIB-dependent system for co-culturing hESCs and hTSCs, which facilitated the construction of human embryoids by aggregating hESCs and hTSCs.
Collapse
Affiliation(s)
- Shuwei Xia
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dainan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yue Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Beijia He
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yin Rong
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Chen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Xiao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Long Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
5
|
Aguila L, Nociti RP, Sampaio RV, Therrien J, Meirelles FV, Felmer RN, Smith LC. Haploid androgenetic development of bovine embryos reveals imbalanced WNT signaling and impaired cell fate differentiation†. Biol Reprod 2023; 109:821-838. [PMID: 37788061 DOI: 10.1093/biolre/ioad124] [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: 02/09/2023] [Revised: 08/09/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
Haploid embryos have contributed significantly to our understanding of the role of parental genomes in development and can be applied to important biotechnology for human and animal species. However, development to the blastocyst stage is severely hindered in bovine haploid androgenetic embryos (hAE). To further our understanding of such developmental arrest, we performed a comprehensive comparison of the transcriptomic profile of morula-stage embryos, which were validated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) of transcripts associated with differentiation in haploid and biparental embryos. Among numerous disturbances, results showed that pluripotency pathways, especially the wingless-related integration site (WNT) signaling, were particularly unbalanced in hAE. Moreover, transcript levels of KLF4, NANOG, POU5F1, SOX2, CDX2, CTNNBL1, AXIN2, and GSK3B were noticeably altered in hAE, suggesting disturbance of pluripotency and canonical WNT pathways. To evaluate the role of WNT on hAE competence, we exposed early Day-5 morula stage embryos to the GSK3B inhibitor CHIR99021. Although no alterations were observed in pluripotency and WNT-related transcripts, exposure to CHIR99021 improved their ability to reach the blastocysts stage, confirming the importance of the WNT pathway in the developmental outcome of bovine hAE.
Collapse
Affiliation(s)
- Luis Aguila
- Centre de Recherche en Reproduction et Fértilité (CRRF), Département de biomédecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Agriculture and Forestry, Universidad de La Frontera, Temuco, Chile
| | - Ricardo P Nociti
- Centre de Recherche en Reproduction et Fértilité (CRRF), Département de biomédecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of Sao Paulo, São Paulo, Brazil
| | - Rafael V Sampaio
- Centre de Recherche en Reproduction et Fértilité (CRRF), Département de biomédecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Jacinthe Therrien
- Centre de Recherche en Reproduction et Fértilité (CRRF), Département de biomédecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Flavio V Meirelles
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of Sao Paulo, São Paulo, Brazil
| | - Ricardo N Felmer
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Agriculture and Forestry, Universidad de La Frontera, Temuco, Chile
| | - Lawrence C Smith
- Centre de Recherche en Reproduction et Fértilité (CRRF), Département de biomédecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| |
Collapse
|
6
|
Ma C, Li Q, Yang Y, Ge L, Cai J, Wang J, Zhu M, Xiong Y, Zhang W, Xie J, Cao Y, Zhao H, Wei Q, Huang C, Shi J, Zhang JV, Duan E, Lei X. mTOR hypoactivity leads to trophectoderm cell failure by enhancing lysosomal activation and disrupting the cytoskeleton in preimplantation embryo. Cell Biosci 2023; 13:219. [PMID: 38037142 PMCID: PMC10688112 DOI: 10.1186/s13578-023-01176-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Metabolic homeostasis is closely related to early impairment of cell fate determination and embryo development. The protein kinase mechanistic target of rapamycin (mTOR) is a key regulator of cellular metabolism in the body. Inhibition of mTOR signaling in early embryo causes postimplantation development failure, yet the mechanisms are still poorly understood. METHODS Pregnancy mice and preimplantation mouse embryo were treated with mTOR inhibitor in vivo and in vitro respectively, and subsequently examined the blastocyst formation, implantation, and post-implantation development. We used immunofluorescence staining, RNA-Seq smart2, and genome-wide bisulfite sequencing technologies to investigate the impact of mTOR inhibitors on the quality, cell fate determination, and molecular alterations in developing embryos. RESULTS We showed mTOR suppression during preimplantation decreases the rate of blastocyst formation and the competency of implantation, impairs the post implantation embryonic development. We discovered that blocking mTOR signaling negatively affected the transformation of 8-cell embryos into blastocysts and caused various deficiencies in blastocyst quality. These included problems with compromised trophectoderm cell differentiation, as well as disruptions in cell fate specification. mTOR suppression significantly affected the transcription and DNA methylation of embryos. Treatment with mTOR inhibitors increase lysosomal activation and disrupts the organization and dynamics of the actin cytoskeleton in blastocysts. CONCLUSIONS These results demonstrate that mTOR plays a crucial role in 8-cell to blastocyst transition and safeguards embryo quality during early embryo development.
Collapse
Affiliation(s)
- Chiyuan Ma
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qin Li
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuxin Yang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou, 571199, China
| | - Lei Ge
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaxuan Cai
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Juan Wang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Maoxian Zhu
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yue Xiong
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenya Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jingtong Xie
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou, 571199, China
| | - Yujing Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huashan Zhao
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qing Wei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chen Huang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junchao Shi
- CAS Key Laboratory of Genome Sciences and Information, China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jian V Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Jing T, Wu Y, Wan A, Ge C, Chen ZJ, Du Y. Circular RNA as a Novel Regulator and Promising Biomarker in Polycystic Ovary Syndrome. Biomolecules 2023; 13:1101. [PMID: 37509138 PMCID: PMC10377156 DOI: 10.3390/biom13071101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/09/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent metabolic and reproductive disorder that causes low fertility in females. Despite its detrimental effects on women's health, care for PCOS has been impeded by its undefined pathogenesis. Thus, there is an urgent need to explore novel biomarkers and therapeutic targets for the diagnosis and treatment of PCOS. Circular RNAs (circRNAs) are a class of noncoding RNAs with covalently closed cyclic structures, present in high abundance, and show development-stage specific expression patterns. Recent studies have demonstrated that circRNAs participate in PCOS progression by modulating various biological functions, including cell proliferation, apoptosis, and steroidogenesis. In addition, circRNAs are widely present in the follicular fluid of women with PCOS, indicating their potential as diagnostic biomarkers and therapeutic targets for PCOS. This review provides the current knowledge of circRNAs in PCOS, including their regulatory functions and molecular mechanisms, and explores their potential as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Tianrui Jing
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yifan Wu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Anran Wan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Chengmin Ge
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250012, China
- NMU-SD Suzhou Collaborative Innovation Center for Reproductive Medicine, Suzhou 215000, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| |
Collapse
|
8
|
Canse C, Yildirim E, Yaba A. Overview of junctional complexes during mammalian early embryonic development. Front Endocrinol (Lausanne) 2023; 14:1150017. [PMID: 37152932 PMCID: PMC10158982 DOI: 10.3389/fendo.2023.1150017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 05/09/2023] Open
Abstract
Cell-cell junctions form strong intercellular connections and mediate communication between blastomeres during preimplantation embryonic development and thus are crucial for cell integrity, polarity, cell fate specification and morphogenesis. Together with cell adhesion molecules and cytoskeletal elements, intercellular junctions orchestrate mechanotransduction, morphokinetics and signaling networks during the development of early embryos. This review focuses on the structure, organization, function and expressional pattern of the cell-cell junction complexes during early embryonic development. Understanding the importance of dynamic junction formation and maturation processes will shed light on the molecular mechanism behind developmental abnormalities of early embryos during the preimplantation period.
Collapse
Affiliation(s)
- Ceren Canse
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Ecem Yildirim
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
| | - Aylin Yaba
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
- *Correspondence: Aylin Yaba,
| |
Collapse
|
9
|
Jeensuk S, Ortega MS, Saleem M, Hawryluk B, Scheffler TL, Hansen PJ. Actions of WNT family member 5A to regulate characteristics of development of the bovine preimplantation embryo†. Biol Reprod 2022; 107:928-944. [PMID: 35765196 PMCID: PMC9562107 DOI: 10.1093/biolre/ioac127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
WNT signaling is important for regulation of embryonic development. The most abundant WNT gene expressed in the bovine endometrium during the preimplantation period is WNT5A. One objective was to determine whether WNT5A regulates competence of the bovine preimplantation embryo to become a blastocyst and alters the number of cells in the inner cell mass and trophectoderm. A second objective was to delineate features of the cell-signaling mechanisms involved in WNT5A actions. WNT5A caused a concentration-dependent increase in the proportion of embryos developing to the blastocyst stage and in the number of inner cell mass cells in the resultant blastocysts. A concentration of 200 ng/mL was most effective, and a higher concentration of 400 ng/mL was not stimulatory. Bovine serum albumin in culture reduced the magnitude of effects of WNT5A on development to the blastocyst stage. WNT5A affected expression of 173 genes at the morula stage; all were upregulated by WNT5A. Many of the upregulated genes were associated with cell signaling. Actions of WNT5A on development to the blastocyst stage were suppressed by a Rho-associated coiled-coil kinase (ROCK) signaling inhibitor, suggesting that WNT5A acts through Ras homology gene family member A (RhoA)/ROCK signaling. Other experiments indicated that actions of WNT5A are independent of the canonical β-catenin signaling pathway and RAC1/c-Jun N-terminal kinase (JNK) signaling. This is the first report outlining the actions of WNT5A to alter the development of the mammalian embryo. These findings provide insights into how embryokines regulate maternal-embryonic communication.
Collapse
Affiliation(s)
- Surawich Jeensuk
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
- Department of Livestock Development, Bureau of Biotechnology in Livestock Production, Pathum Thani, Thailand
| | - M Sofia Ortega
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Muhammad Saleem
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
- Department of Theriogenology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Briana Hawryluk
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Tracy L Scheffler
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Peter J Hansen
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Transcription factor networks in trophoblast development. Cell Mol Life Sci 2022; 79:337. [PMID: 35657505 PMCID: PMC9166831 DOI: 10.1007/s00018-022-04363-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022]
Abstract
The placenta sustains embryonic development and is critical for a successful pregnancy outcome. It provides the site of exchange between the mother and the embryo, has immunological functions and is a vital endocrine organ. To perform these diverse roles, the placenta comprises highly specialized trophoblast cell types, including syncytiotrophoblast and extravillous trophoblast. The coordinated actions of transcription factors (TFs) regulate their emergence during development, subsequent specialization, and identity. These TFs integrate diverse signaling cues, form TF networks, associate with chromatin remodeling and modifying factors, and collectively determine the cell type-specific characteristics. Here, we summarize the general properties of TFs, provide an overview of TFs involved in the development and function of the human trophoblast, and address similarities and differences to their murine orthologs. In addition, we discuss how the recent establishment of human in vitro models combined with -omics approaches propel our knowledge and transform the human trophoblast field.
Collapse
|
11
|
Chronopoulou E, Koika V, Tsiveriotis K, Stefanidis K, Kalogeropoulos S, Georgopoulos N, Adonakis G, Kaponis A. Wnt4, Wnt6 and β-catenin expression in human placental tissue - is there a link with first trimester miscarriage? Results from a pilot study. Reprod Biol Endocrinol 2022; 20:51. [PMID: 35300692 PMCID: PMC8928677 DOI: 10.1186/s12958-022-00923-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 03/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Demystifying the events around early pregnancy is challenging. A wide network of mediators and signaling cascades orchestrate the processes of implantation and trophoblast proliferation. Dysregulation of these pathways could be implicated in early pregnancy loss. There is accumulating evidence around the role of Wnt pathway in implantation and early pregnancy. The purpose of this study was to explore alterations in the expression of Wnt4, Wnt6 and β-catenin in placental tissue obtained from human first trimester euploid miscarriages versus normally developing early pregnancies. METHODS The study group consisted of first trimester miscarriages (early embryonic demises and incomplete miscarriages) and the control group of social terminations of pregnancy (TOPs). The placental mRNA expression of Wnt4, Wnt6 and β-catenin was studied using reverse transcription PCR and real time PCR. Only euploid conceptions were included in the analysis. RESULTS Wnt4 expression was significantly increased in placental tissue from first trimester miscarriages versus controls (p = 0.003). No significant difference was documented in the expression of Wnt6 (p = 0.286) and β-catenin (p = 0.793). There was a 5.1fold increase in Wnt4 expression for early embryonic demises versus TOPs and a 7.6fold increase for incomplete miscarriages versus TOPs - no significant difference between the two subgroups of miscarriage (p = 0.533). CONCLUSIONS This is, to our knowledge, the first study demonstrating significant alteration of Wnt4 expression in human placental tissue, from failed early pregnancies compared to normal controls. Undoubtedly, a more profound study is needed to confirm these preliminary findings and explore Wnt mediators as potential targets for strategies to predict and prevent miscarriage.
Collapse
Affiliation(s)
- Elpiniki Chronopoulou
- Department of Obstetrics and Gynaecology, University General Hospital of Patras, 265 04, Rion, Greece.
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology, University General Hospital of Patras, 265 04, Rion, Greece.
| | - Vasiliki Koika
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology, University General Hospital of Patras, 265 04, Rion, Greece
| | - Konstantinos Tsiveriotis
- Department of Obstetrics and Gynaecology, University General Hospital of Patras, 265 04, Rion, Greece
| | - Konstantinos Stefanidis
- Department of Obstetrics and Gynaecology, University Hospital of Athens, "Alexandra", Lourou 4-2, 115 28, Athens, Greece
| | - Sotirios Kalogeropoulos
- Department of Obstetrics and Gynaecology, University General Hospital of Patras, 265 04, Rion, Greece
| | - Neoklis Georgopoulos
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology, University General Hospital of Patras, 265 04, Rion, Greece
| | - George Adonakis
- Department of Obstetrics and Gynaecology, University General Hospital of Patras, 265 04, Rion, Greece
| | - Apostolos Kaponis
- Department of Obstetrics and Gynaecology, University General Hospital of Patras, 265 04, Rion, Greece
| |
Collapse
|
12
|
Dietrich B, Haider S, Meinhardt G, Pollheimer J, Knöfler M. WNT and NOTCH signaling in human trophoblast development and differentiation. Cell Mol Life Sci 2022; 79:292. [PMID: 35562545 PMCID: PMC9106601 DOI: 10.1007/s00018-022-04285-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/16/2022]
Abstract
Correct development of the human placenta and its differentiated epithelial cells, syncytial trophoblasts (STBs) and extravillous trophoblasts (EVTs), is crucial for a successful pregnancy outcome. STBs develop by cell fusion of mononuclear cytotrophoblasts (CTBs) in placental floating villi, whereas migratory EVTs originate from specialized villi anchoring to the maternal decidua. Defects in trophoblast differentiation have been associated with severe pregnancy disorders such as early-onset preeclampsia and fetal growth restriction. However, the evolutionary pathways underlying normal and adverse placentation are poorly understood. Herein, we discuss Wingless (WNT) and NOTCH signaling, two pathways that play pivotal roles in human placenta and trophoblast development. Whereas WNT is necessary for expansion of trophoblast progenitors and stem cells, NOTCH1 is required for proliferation and survival of EVT precursors. Differentiation of the latter is orchestrated by a switch in NOTCH receptor expression as well as by changes in WNT ligands and their downstream effectors.
Collapse
Affiliation(s)
- Bianca Dietrich
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Sandra Haider
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Gudrun Meinhardt
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Jürgen Pollheimer
- grid.22937.3d0000 0000 9259 8492Maternal-Fetal Immunology Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Martin Knöfler
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| |
Collapse
|
13
|
WNT3 hypomethylation counteracts low activity of the Wnt signaling pathway in the placenta of preeclampsia. Cell Mol Life Sci 2021; 78:6995-7008. [PMID: 34608506 PMCID: PMC8558176 DOI: 10.1007/s00018-021-03941-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/17/2021] [Accepted: 09/13/2021] [Indexed: 01/12/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy. Many studies have shown that epigenetic mechanisms may play a role in preeclampsia. Moreover, our previous study indicated that the differentially methylated genes in preeclampsia were enriched in the Wnt/β-catenin signaling pathway. This study aimed to identify differentially methylated Wnt/β-catenin signaling pathway genes in the preeclamptic placenta and to study the roles of these genes in trophoblast cells in vitro. Using an Illumina Infinium HumanMethylation 850 K BeadChip, we found that the Wnt signaling pathway was globally hypermethylated in the preeclamptic group compared with the term birth group, but hypomethylated in the preeclamptic group compared with the preterm birth group. Among all Wnt/β-catenin signaling pathway factors, WNT3 was the most significantly differentially expressed gene and was hypomethylated in the preeclamptic group compared to the nonhypertensive groups, namely, the preterm birth group and term birth group. This result was confirmed by pyrosequencing. Through quantitative real-time PCR and western blot analysis, the WNT3 gene was found to be highly expressed in preeclamptic placental tissues, in contrast to other WNT factors, which were previously reported to be expressed at low levels in placental tissues. Additionally, in the HTR8/SVneo cell line, knockdown of WNT3 suppressed the Wnt/β-catenin signaling pathway, consistent with the findings for other WNT factors. These results prompted us to speculate that the WNT3 gene counteracts the low activation state of the Wnt signaling pathway in the preeclamptic placenta through methylation modification.
Collapse
|
14
|
Paonessa M, Borini A, Coticchio G. Genetic causes of preimplantation embryo developmental failure. Mol Reprod Dev 2021; 88:338-348. [PMID: 33843124 DOI: 10.1002/mrd.23471] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Embryo development requires orchestrated events, finely regulated at the molecular and cellular level by mechanisms which are progressively emerging from animal studies. With progress in genetic technologies-such as genome editing and single-cell RNA analysis-we can now assess embryo gene expression with increased precision and gain new insights into complex processes until recently difficult to explore. Multiple genes and regulative pathways have been identified for each developmental stage. We have learned that embryos with undisturbed and timely gene expression have higher chances of successful development. For example, selected genes are highly expressed during the first stages, being involved in cell adhesion, cell cycle, and regulation of transcription; other genes are instead crucial for lineage specification and therefore expressed at later stages. Due to ethical constraints, studies on human embryos remain scarce, mainly descriptive, and unable to provide functional evidence. This highlights the importance of animal studies as basic knowledge to test and appraise in a clinical context. In this review, we report on preimplantation development with a focus on genes whose impairment leads to developmental arrest. Preconceptional genetic screening could identify loss-of-function mutations of these genes; thereby, novel biomarkers of embryo quality could be adopted to improve diagnosis and treatment of infertility.
Collapse
Affiliation(s)
- Mariagrazia Paonessa
- 9.Baby, Family and Fertility Center, Bologna, Italy.,Casa di Cura Candela Spa, Palermo, Italy
| | | | | |
Collapse
|
15
|
Hu Y, Huang K, Zeng Q, Feng Y, Ke Q, An Q, Qin LJ, Cui Y, Guo Y, Zhao D, Peng Y, Tian D, Xia K, Chen Y, Ni B, Wang J, Zhu X, Wei L, Liu Y, Xiang P, Liu JY, Xue Z, Fan G. Single-cell analysis of nonhuman primate preimplantation development in comparison to humans and mice. Dev Dyn 2021; 250:974-985. [PMID: 33449399 DOI: 10.1002/dvdy.295] [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: 07/23/2020] [Revised: 12/14/2020] [Accepted: 12/19/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genetic programs underlying preimplantation development and early lineage segregation are highly conserved across mammals. It has been suggested that nonhuman primates would be better model organisms for human embryogenesis, but a limited number of studies have investigated the monkey preimplantation development. In this study, we collect single cells from cynomolgus monkey preimplantation embryos for transcriptome profiling and compare with single-cell RNA-seq data derived from human and mouse embryos. RESULTS By weighted gene-coexpression network analysis, we found that cynomolgus gene networks have greater conservation with human embryos including a greater number of conserved hub genes than that of mouse embryos. Consistently, we found that early ICM/TE lineage-segregating genes in monkeys exhibit greater similarity with human when compared to mouse, so are the genes in signaling pathways such as LRP1 and TCF7 involving in WNT pathway. Last, we tested the role of one conserved pre-EGA hub gene, SIN3A, using a morpholino knockdown of maternal RNA transcripts in monkey embryos followed by single-cell RNA-seq. We found that SIN3A knockdown disrupts the gene-silencing program during the embryonic genome activation transition and results in developmental delay of cynomolgus embryos. CONCLUSION Taken together, our study provided new insight into evolutionarily conserved and divergent transcriptome dynamics during mammalian preimplantation development.
Collapse
Affiliation(s)
- Youjin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun-Ye-Sat University, Guangzhou, China.,Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Kevin Huang
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Qiao Zeng
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yun Feng
- Reproductive Medicine Center, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, China
| | - Qiong Ke
- Key Laboratory of Stem Cell Engineering Ministry of Education, Zhongshan College of Medicine, Sun-Ye-Sat University, Guangzhou, China
| | - Qin An
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Lian-Ju Qin
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - YuGui Cui
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Guo
- The Second Affiliated Hospital, Xiangya School of Medicine, Central South University, Changsha, China
| | - Dicheng Zhao
- State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu Peng
- State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, China
| | - Di Tian
- State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, China
| | - Kun Xia
- State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yong Chen
- Key Laboratory of Genetics and Birth Health of Hunan Province, Changsha, China
| | - Bin Ni
- Key Laboratory of Genetics and Birth Health of Hunan Province, Changsha, China
| | - Jinmei Wang
- Shanghai East Hospital, School of Life Sciences & Technology, Tongji University, Shanghai, China
| | - Xianmin Zhu
- Shanghai East Hospital, School of Life Sciences & Technology, Tongji University, Shanghai, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun-Ye-Sat University, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun-Ye-Sat University, Guangzhou, China
| | - Peng Xiang
- Key Laboratory of Stem Cell Engineering Ministry of Education, Zhongshan College of Medicine, Sun-Ye-Sat University, Guangzhou, China
| | - Jia-Yin Liu
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhigang Xue
- Reproductive Medicine Center, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, China
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
16
|
Kaloğlu C, Bulut HE, Hamutoğlu R, Korkmaz EM, Önder O, Dağdeviren T, Aydemir MN. Wingless ligands and beta-catenin expression in the rat endometrium: The role of Wnt3 and Wnt7a/beta-catenin pathway at the embryo-uterine interface. Mol Reprod Dev 2020; 87:1159-1172. [PMID: 32949181 DOI: 10.1002/mrd.23423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 11/05/2022]
Abstract
Wnt/beta-catenin signaling may play an essential role in endometrial decidualization, placentation, and the establishment of pregnancy. We investigate here the possible roles, immunolocalizations, and synthesis of the Wnt3, Wnt7a, and beta-catenin proteins in the rat endometrium during the estrous cycle and early postimplantation period. Wnt3 and Wnt7a had a similar localization and dynamic expression relative to the endometrial stages. Wnt7a immunostaining was not limited only to the luminal epithelial cells, but also to strong stainings in the stromal and endothelial cells. Wnt3, Wnt7a, and beta-catenin were highly synthesized and colocalized at the trophoblast-decidual interface; and were more obvious in the primary decidual zone, the GTCs, and the ectoplacental cone. Beta-catenin was strongly localized at the borders of the mature decidual cells; however, Wnt3 and Wnt7a immunolocalizations were decreased in those cells. As such, the immunolocalization of Wnt3, Wnt7a, and beta-catenin shifted with decidualization and placentation. The expression level of Wnt3, Wnt7a, and beta-catenin messenger RNAs increased in early pregnancy, and especially between Days 8.5 and 9.5. The dramatic changes in the expression of Wnt3, Wnt7a, and beta-catenin observed during the early days of pregnancy and the estrous cycle may indicate their roles in decidualization, stromal cell proliferation, and trophoblast invasion.
Collapse
Affiliation(s)
- Celal Kaloğlu
- Assisted Reproduction Technology (ART) Center, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey.,Department of Histology and Embryology, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Hüseyin E Bulut
- Department of Histology and Embryology, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Rasim Hamutoğlu
- Department of Histology and Embryology, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Ertan M Korkmaz
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Ozan Önder
- Department of Histology and Embryology, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Tuğba Dağdeviren
- Department of Histology and Embryology, Faculty of Medicine, Sivas-Cumhuriyet University, Sivas, Turkey
| | - Merve N Aydemir
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas-Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
17
|
Płusa B, Piliszek A. Common principles of early mammalian embryo self-organisation. Development 2020; 147:147/14/dev183079. [PMID: 32699138 DOI: 10.1242/dev.183079] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pre-implantation mammalian development unites extreme plasticity with a robust outcome: the formation of a blastocyst, an organised multi-layered structure ready for implantation. The process of blastocyst formation is one of the best-known examples of self-organisation. The first three cell lineages in mammalian development specify and arrange themselves during the morphogenic process based on cell-cell interactions. Despite decades of research, the unifying principles driving early mammalian development are still not fully defined. Here, we discuss the role of physical forces, and molecular and cellular mechanisms, in driving self-organisation and lineage formation that are shared between eutherian mammals.
Collapse
Affiliation(s)
- Berenika Płusa
- Faculty of Biology, Medicine and Health (FBMH), Division of Developmental Biology & Medicine, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Anna Piliszek
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Postepu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
18
|
Li X, Zhang W, Fu J, Xu Y, Gu R, Qu R, Li L, Sun Y, Sun X. MicroRNA-451 is downregulated in the follicular fluid of women with endometriosis and influences mouse and human embryonic potential. Reprod Biol Endocrinol 2019; 17:96. [PMID: 31744497 PMCID: PMC6862852 DOI: 10.1186/s12958-019-0538-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous work demonstrated that there are numerous miRNAs in human follicular fluids, some of which are associated with reproductive diseases. In the current study, we sought to determine whether microRNAs (miRNAs) in the follicular fluid (FF) are differentially expressed between women with and without endometriosis and to uncover the association of miRNAs with the oocyte and embryonic development potential. METHODS FF was harvested from 30 women with endometriosis and 30 women without who underwent in vitro fertilization treatment at the University Hospital between February and December 2016. The FF samples were subjected to miRNA profiling and validation via quantitative reverse transcription polymerase chain reaction analysis. Mouse/human metaphase-I (MI) oocytes were harvested and micro-injected with an miR-451 inhibitor, and the effects of miR-451 knockdown on Wnt/WNT signalling genes were investigated. RESULTS Oocyte number, fertilization rate, and number of available embryos were decreased significantly in women with endometriosis relative to those without endometriosis. Hsa-miR-451 in FF was downregulated in endometriosis patients relative to control subjects (P < 0.01). Moreover, the proportions of mouse/human MI oocytes that developed into 2-pronuclei (2PN), 2-cell, 8-10-cell and blastocyst-stage embryos were affected by miR-451 knockdown in mouse/human oocytes. Components of the Wnt signalling pathway were aberrantly expressed in the mouse/human oocytes and embryos in the miR-451 inhibitor-injected group. CONCLUSIONS miR-451 was downregulated in FF samples from endometriosis patients and was modestly effective in distinguishing endometriosis patients from non-endometriosis patients. miR-451 downregulation in mouse and human oocytes affected pre-implantation embryogenesis by suppressing the Wnt signalling pathway. This miRNA might serve as a novel biomarker of oocyte and embryo quality in assisted reproductive treatment.
Collapse
Affiliation(s)
- Xiong Li
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Wenbi Zhang
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Jing Fu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yan Xu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Ruihuan Gu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Ronggui Qu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Lu Li
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yijuan Sun
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
- Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
19
|
Wu J, Feng X, Du Y, Luan B, Yu H, Yu Y, Wu L, Zhao H. β-catenin/LIN28B promotes the proliferation of human choriocarcinoma cells via Let-7a repression. Acta Biochim Biophys Sin (Shanghai) 2019; 51:455-462. [PMID: 30958882 DOI: 10.1093/abbs/gmz027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 01/03/2023] Open
Abstract
Choriocarcinoma is a rare and malignant trophoblastic tumor. However, the molecular mechanisms by which choriocarcinoma is regulated remain unknown. In the present study, we first elucidated that LIN28B was highly expressed in human choriocarcinoma tissues and choriocarcinoma cell lines. Our data further demonstrated that knockdown of LIN28B by small interfering RNA caused an increase in Let-7a expression in JAR cells. In addition, silencing of LIN28B inhibited IGF2BP1 expression and suppressed cell proliferation capacity, both of which can be markedly restored by Let-7a inhibitor. In contrast, LIN28B over-expression-improved cell proliferation was inhibited by Let-7a mimic. Knockdown of β-catenin resulted in reduced expression of LIN28B and increased expression of Let-7a. Knockdown of β-catenin also caused a decrease in cell proliferation, which can be recovered by re-expression of LIN28B or by Let-7a inhibitor. Collectively, our data indicate that β-catenin/LIN28B/Let-7a pathway may be crucial for the regulation of cell proliferation in human choriocarcinoma cells.
Collapse
Affiliation(s)
- Jing Wu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xuan Feng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yan Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
| | - Baoxin Luan
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Huandi Yu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yinhua Yu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Lanxiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Hongbo Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
20
|
Wang C, Han X, Zhou Z, Uyunbilig B, Huang X, Li R, Li X. Wnt3a Activates the WNT-YAP/TAZ Pathway to Sustain CDX2 Expression in Bovine Trophoblast Stem Cells. DNA Cell Biol 2019; 38:410-422. [DOI: 10.1089/dna.2018.4458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Chen Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestocks, Inner Mongolia University, Hohhot, China
| | - Xuejie Han
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestocks, Inner Mongolia University, Hohhot, China
| | - Zhengwei Zhou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestocks, Inner Mongolia University, Hohhot, China
| | - Borjigin Uyunbilig
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestocks, Inner Mongolia University, Hohhot, China
| | - Xianghua Huang
- Department of Urology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Rongfeng Li
- State Key Laboratories of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Xueling Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestocks, Inner Mongolia University, Hohhot, China
| |
Collapse
|
21
|
Tribulo P, Leão BCDS, Lehloenya KC, Mingoti GZ, Hansen PJ. Consequences of endogenous and exogenous WNT signaling for development of the preimplantation bovine embryo. Biol Reprod 2018; 96:1129-1141. [PMID: 28575156 PMCID: PMC5803770 DOI: 10.1093/biolre/iox048] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023] Open
Abstract
The specific role of WNT signaling during preimplantation development remains unclear. Here, we evaluated consequences of activation and inhibition of β-catenin (CTNNB1)-dependent and -independent WNT signaling in the bovine preimplantation embryo. Activation of CTNNB1-mediated WNT signaling by the agonist 2-amino-4-(3,4-(methylenedioxy)benzylamino)-6-(3-methoxyphenyl)pyrimidine (AMBMP) and a glycogen synthase kinase 3 inhibitor reduced development to the blastocyst stage. Moreover, the antagonist of WNT signaling, dickkopf-related protein 1 (DKK1), alleviated the negative effect of AMBMP on development via reduction of CTNNB1. Based on labeling for phospho c-Jun N-terminal kinase, there was no evidence that DKK1 activated the planar cell polarity (PCP) pathway. Inhibition of secretion of endogenous WNTs did not affect development but increased number of cells in the inner cell mass (ICM). In contrast, DKK1 did not affect number of ICM or trophectoderm (TE) cells, suggesting that embryo-derived WNTs regulate ICM proliferation through a mechanism independent of CTNNB1. In addition, DKK1 did not affect the number of cells positive for the transcription factor yes-associated protein 1 (YAP1) involved in TE formation. In fact, DKK1 decreased YAP1. In contrast, exposure of embryos to WNT family member 7A (WNT7A) improved blastocyst development, inhibited the PCP pathway, and did not affect amounts of CTNNB1. Results indicate that embryo-derived WNTs are dispensable for blastocyst formation but participate in regulation of ICM proliferation, likely through a mechanism independent of CTNNB1. The response to AMBMP and WNT7A leads to the hypothesis that maternally derived WNTs can play a positive or negative role in regulation of preimplantation development.
Collapse
Affiliation(s)
- Paula Tribulo
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Beatriz Caetano da Silva Leão
- School of Veterinary Medicine, Laboratory of Reproductive Physiology, UNESP-Universidade Estadual Paulista, Araçatuba, São Paulo, Brazil and Post-Graduation Program in Veterinary Medicine, School of Agrarian and Veterinarian Sciences, Department of Animal Reproduction, UNESP-Universidade Estadual Paulista, Jaboticabal, São Paulo, Brazil
| | - Khoboso C Lehloenya
- Department of Animal and Wildlife Sciences, University of Pretoria, Pretoria, South Africa
| | - Gisele Zoccal Mingoti
- School of Veterinary Medicine, Laboratory of Reproductive Physiology, UNESP-Universidade Estadual Paulista, Araçatuba, São Paulo, Brazil and Post-Graduation Program in Veterinary Medicine, School of Agrarian and Veterinarian Sciences, Department of Animal Reproduction, UNESP-Universidade Estadual Paulista, Jaboticabal, São Paulo, Brazil
| | - Peter J Hansen
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
22
|
The Pleiotropic Effects of the Canonical Wnt Pathway in Early Development and Pluripotency. Genes (Basel) 2018; 9:genes9020093. [PMID: 29443926 PMCID: PMC5852589 DOI: 10.3390/genes9020093] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Abstract
The technology to derive embryonic and induced pluripotent stem cells from early embryonic stages and adult somatic cells, respectively, emerged as a powerful resource to enable the establishment of new in vitro models, which recapitulate early developmental processes and disease. Additionally, pluripotent stem cells (PSCs) represent an invaluable source of relevant differentiated cell types with immense potential for regenerative medicine and cell replacement therapies. Pluripotent stem cells support self-renewal, potency and proliferation for extensive periods of culture in vitro. However, the core pathways that rule each of these cellular features specific to PSCs only recently began to be clarified. The Wnt signaling pathway is pivotal during early embryogenesis and is central for the induction and maintenance of the pluripotency of PSCs. Signaling by the Wnt family of ligands is conveyed intracellularly by the stabilization of β-catenin in the cytoplasm and in the nucleus, where it elicits the transcriptional activity of T-cell factor (TCF)/lymphoid enhancer factor (LEF) family of transcription factors. Interestingly, in PSCs, the Wnt/β-catenin–TCF/LEF axis has several unrelated and sometimes opposite cellular functions such as self-renewal, stemness, lineage commitment and cell cycle regulation. In addition, tight control of the Wnt signaling pathway enhances reprogramming of somatic cells to induced pluripotency. Several recent research efforts emphasize the pleiotropic functions of the Wnt signaling pathway in the pluripotent state. Nonetheless, conflicting results and unanswered questions still linger. In this review, we will focus on the diverse functions of the canonical Wnt signaling pathway on the developmental processes preceding embryo implantation, as well as on its roles in pluripotent stem cell biology such as self-renewal and cell cycle regulation and somatic cell reprogramming.
Collapse
|
23
|
Tríbulo P, Siqueira L, Oliveira L, Scheffler T, Hansen P. Identification of potential embryokines in the bovine reproductive tract. J Dairy Sci 2018; 101:690-704. [DOI: 10.3168/jds.2017-13221] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022]
|
24
|
Ramos-Ibeas P, Nichols J, Alberio R. States and Origins of Mammalian Embryonic Pluripotency In Vivo and in a Dish. Curr Top Dev Biol 2017; 128:151-179. [PMID: 29477162 DOI: 10.1016/bs.ctdb.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mouse embryonic stem cells (ESC), derived from preimplantation embryos in 1981, defined mammalian pluripotency for many decades. However, after the derivation of human ESC in 1998, comparative studies showed that different types of pluripotency exist in early embryos and that these can be captured in vitro under various culture conditions. Over the past decade much has been learned about the key signaling pathways, growth factor requirements, and transcription factor profiles of pluripotent cells in embryos, allowing improvement of derivation and culture conditions for novel pluripotent stem cell types. More recently, studies using single-cell transcriptomics of embryos from different species provided an unprecedented level of resolution of cellular interactions and cell fate decisions that are informing new ways to understand the emergence of pluripotency in different organisms. These new approaches enhance knowledge of species differences during early embryogenesis and will be instrumental for improving methodologies for generating intra- and interspecies chimeric animals using pluripotent stem cells. Here, we discuss the recent developments in our understanding of early embryogenesis in different mammalian species.
Collapse
Affiliation(s)
| | - Jennifer Nichols
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; University of Cambridge, Cambridge, United Kingdom.
| | - Ramiro Alberio
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
25
|
Zhao H, Wu L, Wu J, Yu H, Zhou J, Luan B, Xu C. Aberrantly Expressed SALL4 Promotes Cell Proliferation via β-Catenin/c-Myc Pathway in Human Choriocarcinoma Cells. Reprod Sci 2017. [DOI: 10.1177/1933719117715130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Hongbo Zhao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Lanxiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jing Wu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Huandi Yu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jiayi Zhou
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Baoxin Luan
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Congjian Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Zhang Z, Wang X, Zhang L, Shi Y, Wang J, Yan H. Wnt/β-catenin signaling pathway in trophoblasts and abnormal activation in preeclampsia (Review). Mol Med Rep 2017; 16:1007-1013. [PMID: 29067442 DOI: 10.3892/mmr.2017.6718] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/13/2017] [Indexed: 11/06/2022] Open
Abstract
Preeclampsia (PE) is one of the most common types of hypertensive disease and occurs in 3‑4% of pregnancies. There are a number of theories on the pathogenesis of PE. Abnormal differentiation of the placenta may lead to failure of trophoblast migration, shallow placenta implantation and placental ischemia/hypoxia, followed by the subsequent occurrence of PE. The Wnt/β-catenin pathway is a canonical Wnt‑signaling pathway that regulates several biological processes, including proliferation, migration, invasion and apoptosis. Abnormal activation of the Wnt/β‑catenin signaling pathway may serve an important role in the pathogenesis of various human diseases, particularly in human cancer. Recent studies have demonstrated that the dysregulation of the Wnt/β‑catenin signaling pathway may contribute to PE. The present review aims to summarize the articles on Wnt/β‑catenin signaling pathway in the trophoblast and abnormal activation in PE. Wnt/β-catenin signaling may serve a significant role in the pathogenesis of PE and may be a prospective therapeutic target for the prevention and treatment of PE.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaofang Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Linlin Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ying Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jinming Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huan Yan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
27
|
Wu J, Izpisua Belmonte JC. Stem Cells: A Renaissance in Human Biology Research. Cell 2017; 165:1572-1585. [PMID: 27315475 DOI: 10.1016/j.cell.2016.05.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Indexed: 12/18/2022]
Abstract
The understanding of human biology and how it relates to that of other species represents an ancient quest. Limited access to human material, particularly during early development, has restricted researchers to only scratching the surface of this inherently challenging subject. Recent technological innovations, such as single cell "omics" and human stem cell derivation, have now greatly accelerated our ability to gain insights into uniquely human biology. The opportunities afforded to delve molecularly into scarce material and to model human embryogenesis and pathophysiological processes are leading to new insights of human development and are changing our understanding of disease and choice of therapy options.
Collapse
Affiliation(s)
- Jun Wu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, 135, Guadalupe 30107, Murcia, Spain
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
28
|
Tribulo P, Moss JI, Ozawa M, Jiang Z, Tian XC, Hansen PJ. WNT regulation of embryonic development likely involves pathways independent of nuclear CTNNB1. Reproduction 2017; 153:405-419. [PMID: 28069902 DOI: 10.1530/rep-16-0610] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/07/2016] [Accepted: 01/09/2017] [Indexed: 12/24/2022]
Abstract
The bovine was used to examine the potential for WNT signaling to affect the preimplantation embryo. Expression of seven key genes involved in canonical WNT signaling declined to a nadir at the morula or blastocyst stage. Expression of 80 genes associated with WNT signaling in the morula and inner cell mass (ICM) and trophectoderm (TE) of the blastocyst was also evaluated. Many genes associated with WNT signaling were characterized by low transcript abundance. Seven genes were different between ICM and TE, and all of them were overexpressed in TE as compared to ICM, including WNT6, FZD1, FZD7, LRP6, PORCN, APC and SFRP1 Immunoreactive CTNNB1 was localized primarily to the plasma membrane at all stages examined from the 2-cell to blastocyst stages of development. Strikingly, neither CTNNB1 nor non-phospho (i.e., active) CTNNB1 was observed in the nucleus of blastomeres at any stage of development even after the addition of WNT activators to culture. In contrast, CTNNB1 associated with the plasma membrane was increased by activators of WNT signaling. The planar cell polarity pathway (PCP) could be activated in the embryo as indicated by an experiment demonstrating an increase in phospho-JNK in the nucleus of blastocysts treated with the non-canonical WNT11. Furthermore, WNT11 improved development to the blastocyst stage. In conclusion, canonical WNT signaling is attenuated in the preimplantation bovine embryo but WNT can activate the PCP component JNK. Thus, regulation of embryonic development by WNT is likely to involve activation of pathways independent of nuclear actions of CTNNB1.
Collapse
Affiliation(s)
- Paula Tribulo
- Department of Animal SciencesD.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - James I Moss
- Department of Animal SciencesD.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Manabu Ozawa
- Laboratory of Developmental GeneticsInstitute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Zongliang Jiang
- Center for Regenerative BiologyDepartment of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Xiuchun Cindy Tian
- Center for Regenerative BiologyDepartment of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Peter J Hansen
- Department of Animal SciencesD.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
29
|
Baines K, Renaud S. Transcription Factors That Regulate Trophoblast Development and Function. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 145:39-88. [DOI: 10.1016/bs.pmbts.2016.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
30
|
Novel O-GlcNAcylation on Ser(40) of canonical H2A isoforms specific to viviparity. Sci Rep 2016; 6:31785. [PMID: 27615797 PMCID: PMC5018834 DOI: 10.1038/srep31785] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 07/26/2016] [Indexed: 11/23/2022] Open
Abstract
We report here newly discovered O-linked-N-acetylglucosamine (O-GlcNAc) modification of histone H2A at Ser40 (H2AS40Gc). The mouse genome contains 18 H2A isoforms, of which 13 have Ser40 and the other five have Ala40. The combination of production of monoclonal antibody and mass spectrometric analyses with reverse-phase (RP)-high performance liquid chromatography (HPLC) fractionation indicated that the O-GlcNAcylation is specific to the Ser40 isoforms. The H2AS40Gc site is in the L1 loop structure where two H2A molecules interact in the nucleosome. Targets of H2AS40Gc are distributed genome-wide and are dramatically changed during the process of differentiation in mouse trophoblast stem cells. In addition to the mouse, H2AS40Gc was also detected in humans, macaques and cows, whereas non-mammalian species possessing only the Ala40 isoforms, such as silkworms, zebrafish and Xenopus showed no signal. Genome database surveys revealed that Ser40 isoforms of H2A emerged in Marsupialia and persisted thereafter in mammals. We propose that the emergence of H2A Ser40 and its O-GlcNAcylation linked a genetic event to genome-wide epigenetic events that correlate with the evolution of placental animals.
Collapse
|
31
|
Expression of Wnt/β-Catenin Signaling Pathway and Its Regulatory Role in Type I Collagen with TGF-β1 in Scleral Fibroblasts from an Experimentally Induced Myopia Guinea Pig Model. J Ophthalmol 2016; 2016:5126560. [PMID: 27247798 PMCID: PMC4877496 DOI: 10.1155/2016/5126560] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/18/2016] [Indexed: 11/24/2022] Open
Abstract
Background. To investigate Wnt/β-catenin signaling pathway expression and its regulation of type I collagen by TGF-β1 in scleral fibroblasts from form-deprivation myopia (FDM) guinea pig model. Methods. Wnt isoforms were examined using genome microarrays. Scleral fibroblasts from FDM group and self-control (SC) group were cultured. Wnt isoforms, β-catenin, TGF-β1, and type I collagen expression levels were examined in the two groups with or without DKK-1 or TGF-β1 neutralizing antibody. Results. For genome microarrays, the expression of Wnt3 in FDM group was significantly greater as confirmed in retinal and scleral tissue. The expression of Wnt3 and β-catenin significantly increased in FDM group and decreased significantly with DKK-1. TGF-β1 expression level decreased significantly in FDM group and increased significantly with DKK-1. Along with morphological misalignment inside and outside cells, the amount of type I collagen decreased in FDM group. Furthermore, type I collagen increased and became regular in DKK-1 intervention group, whereas it decreased and rearranged more disorder in TGF-β1 neutralizing antibody intervention group. Conclusions. The activation of Wnt3/β-catenin signaling pathway was demonstrated in primary scleral fibroblasts in FDM. This pathway further reduced the expression of type I collagen by TGF-β1, which ultimately played a role in scleral remodeling during myopia development.
Collapse
|
32
|
Piliszek A, Grabarek JB, Frankenberg SR, Plusa B. Cell fate in animal and human blastocysts and the determination of viability. Mol Hum Reprod 2016; 22:681-690. [DOI: 10.1093/molehr/gaw002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/08/2016] [Indexed: 12/25/2022] Open
|