1
|
El-Sheikh M, Mesalam AA, El-Sayed AF, Mesalam A, Metwally HM, Lee SH, Kong IK. Nicotinamide dual treatment enhances morula-to-blastocyst transition through binding to Zonula Occludens-1 protein. Theriogenology 2025; 237:110-119. [PMID: 39987859 DOI: 10.1016/j.theriogenology.2025.02.022] [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: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Nicotinamide (NAM) is a widely utilized compound in cell culture systems, yet its role during the morula-to-blastocyst transition remains underexplored. This study investigates the effects of NAM supplementation during both in vitro maturation (IVM) of oocytes and late-stage embryo culture (IVC3; the morula stage) on blastocyst development, metabolic flux, mitochondrial bioenergetics, and pluripotency of cells. Bovine oocytes were subjected to dual NAM treatment during IVM and IVC3 and its impact was assessed through cleavage and blastocyst development rates, mitochondrial membrane potential (ΔΨm), and the expression of key metabolic and pluripotency markers using RT-qPCR and immunofluorescence. Additionally, molecular docking was performed to evaluate NAM's interaction with Zonula Occludens-1 (ZO-1) protein. Dual NAM administration significantly increased both blastocyst formation and hatching rates. Computational modeling revealed a strong binding affinity (-6.44 kcal/mol) between NAM and the ZO-1 protein, associated with the morula-to-blastocyst transition. Quantitative RT-PCR analysis showed upregulation of genes related to NAD + biosynthesis (NAMPT, MDH1), glycolysis (PFK1), glycogenesis (GSK-3A), and mitochondrial bioenergetics (SDHA, ND2, ATPase8, TFAM) in NAM-treated group. Additionally, mitochondrial profiling demonstrated enhanced polarization, and OCT4 expression was elevated in NAM-treated embryos. These findings underscore NAM's potential role in enhancing morula-to-blastocyst transition, improving embryonic development through metabolic and mitochondrial regulation, as well as pluripotency factor enhancement.
Collapse
Affiliation(s)
- Marwa El-Sheikh
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre (NRC), Cairo, 12622, Egypt.
| | - Ahmed Atef Mesalam
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, 12622, Egypt.
| | - Ahmed F El-Sayed
- Microbial Genetics Department, Biotechnology Research Institute, National Research Centre, Cairo, 12622, Egypt; Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt.
| | - Ayman Mesalam
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Heba M Metwally
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt.
| | - Seo-Hyun Lee
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju, 52828, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; The King Kong Corp. Ltd., Gyeongsang National University, Jinju, 52828, Republic of Korea
| |
Collapse
|
2
|
Peng B, Wang Q, Zhang F, Shen H, Du P. Mouse totipotent blastomere-like cells model embryogenesis from zygotic genome activation to post implantation. Cell Stem Cell 2025; 32:391-408.e11. [PMID: 39826539 DOI: 10.1016/j.stem.2024.12.006] [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: 09/15/2023] [Revised: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
Embryo development begins with zygotic genome activation (ZGA), eventually generating blastocysts for implantation. However, in vitro systems modeling the pre-implantation development are still absent and challenging. Here, we used mouse totipotent blastomere-like cells (TBLCs) to develop spontaneous differentiation and blastoid formation systems, respectively. We found Wnt signaling enabled the rapid expansion of TBLCs and the optimization of their culture medium. We successfully developed a TBLC-spontaneous differentiation system in which mouse TBLCs (mTBLCs) firstly converted into two types of ZGA-like cells (ZLCs) distinguished by Zscan4 expression. Surprisingly, Zscan4-, but not Zscan4+, ZLCs further passed through intermediate 4-cell and then 8-cell/morula stages to produce epiblast, primitive endoderm, and trophectoderm lineages. Significantly, single TBLCs underwent expansion, compaction, and polarization to efficiently generate blastocyst-like structures and even post-implantation egg-cylinder-like structures. Conclusively, we established TBLC-based differentiation and embryo-like structure formation systems to model early embryonic development, offering criteria for evaluating and understanding totipotency.
Collapse
Affiliation(s)
- Bing Peng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Qingyi Wang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feixiang Zhang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Shen
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Peng Du
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Center of RNA Biology, Peking University, Beijing 100871, China.
| |
Collapse
|
3
|
Huang H, Gao S, Bao M. Exploring Mechanical Forces Shaping Self-Organization and Morphogenesis During Early Embryo Development. Annu Rev Cell Dev Biol 2024; 40:75-96. [PMID: 38608312 DOI: 10.1146/annurev-cellbio-120123-105748] [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] [Indexed: 04/14/2024]
Abstract
Embryonic development is a dynamic process orchestrated by a delicate interplay of biochemical and biophysical factors. While the role of genetics and biochemistry in embryogenesis has been extensively studied, recent research has highlighted the significance of mechanical regulation in shaping and guiding this intricate process. Here, we provide an overview of the current understanding of the mechanical regulation of embryo development. We explore how mechanical forces generated by cells and tissues play a crucial role in driving the development of different stages. We examine key morphogenetic processes such as compaction, blastocyst formation, implantation, and egg cylinder formation, and discuss the mechanical mechanisms and cues involved. By synthesizing the current body of literature, we highlight the emerging concepts and open questions in the field of mechanical regulation. We aim to provide an overview of the field, inspiring future investigations and fostering a deeper understanding of the mechanical aspects of embryo development.
Collapse
Affiliation(s)
- Hong Huang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China;
| | - Min Bao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| |
Collapse
|
4
|
Miki R, Matsuo S, Ushida T, Tano S, Imai K, Nawa A, Kajiyama H, Kotani T. TJP1 suppresses trophoblast cell invasion by expressing E2F8 in the human placenta. Mol Cell Endocrinol 2024; 591:112277. [PMID: 38795825 DOI: 10.1016/j.mce.2024.112277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Adequate extravillous trophoblast (EVT) invasion into the maternal decidua is important for human placental development. We identified that E2F transcription factor 8 (E2F8) suppresses EVT invasion, and that tight junction protein-1 (TJP1) is a potential downstream target gene of E2F8. We investigated the role of TJP1 in the human placenta and regulation of TJP1 expression by E2F8. TJP1 expression decreased in E2F8 knockdown HTR-8/SVneo cells. TJP1 and E2F8 were co-expressed in villi in the first-trimester placenta and in EVTs and villi in the third-trimester placenta. TJP1 was significantly increased in the pre-eclamptic compared with control placenta. TJP1 knockdown increased the invasion of HTR-8/SVneo cells, while TJP1 overexpression inhibited cell invasion. Halo-E2F8 overexpression significantly increased TJP1 expression and TJP1 transcription compared with control placenta. Our findings suggest that E2F8 promotes TJP1 transcription, and that TJP1 expression by E2F8 inhibits EVT invasion. TJP1 and E2F8 may be related to pre-eclampsia pathogenesis.
Collapse
Affiliation(s)
- Rika Miki
- Laboratory of Bell Research Center Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| | - Seiko Matsuo
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan; Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466-8560, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Akihiro Nawa
- Laboratory of Bell Research Center Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan; Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466-8560, Japan.
| |
Collapse
|
5
|
Nakagawa S, Carnevali D, Tan X, Alvarez MJ, Parfitt DE, Di Vicino U, Arumugam K, Shin W, Aranda S, Normanno D, Sebastian-Perez R, Cannatá C, Cortes P, Neguembor MV, Shen MM, Califano A, Cosma MP. The Wnt-dependent master regulator NKX1-2 controls mouse pre-implantation development. Stem Cell Reports 2024; 19:689-709. [PMID: 38701778 PMCID: PMC11103935 DOI: 10.1016/j.stemcr.2024.04.004] [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: 06/23/2022] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Embryo size, specification, and homeostasis are regulated by a complex gene regulatory and signaling network. Here we used gene expression signatures of Wnt-activated mouse embryonic stem cell (mESC) clones to reverse engineer an mESC regulatory network. We identify NKX1-2 as a novel master regulator of preimplantation embryo development. We find that Nkx1-2 inhibition reduces nascent RNA synthesis, downregulates genes controlling ribosome biogenesis, RNA translation, and transport, and induces severe alteration of nucleolus structure, resulting in the exclusion of RNA polymerase I from nucleoli. In turn, NKX1-2 loss of function leads to chromosome missegregation in the 2- to 4-cell embryo stages, severe decrease in blastomere numbers, alterations of tight junctions (TJs), and impairment of microlumen coarsening. Overall, these changes impair the blastocoel expansion-collapse cycle and embryo cavitation, leading to altered lineage specification and developmental arrest.
Collapse
Affiliation(s)
- Shoma Nakagawa
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Davide Carnevali
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Xiangtian Tan
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Mariano J Alvarez
- Department of Systems Biology, Columbia University, New York, NY, USA; DarwinHealth Inc, New York, NY, USA
| | - David-Emlyn Parfitt
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Umberto Di Vicino
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Karthik Arumugam
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - William Shin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Sergi Aranda
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Davide Normanno
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Institute of Human Genetics, CNRS, Montpellier, France
| | - Ruben Sebastian-Perez
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Chiara Cannatá
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Paola Cortes
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Maria Victoria Neguembor
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Michael M Shen
- Department of Systems Biology, Columbia University, New York, NY, USA; Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Chan Zuckerberg Biohub New York, New York, NY, USA.
| | - Maria Pia Cosma
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; ICREA, Pg.Lluis Companys 23, 08010 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Yuexiu District, Guangzhou 510080, China.
| |
Collapse
|
6
|
Fang RY, Liu YW, Goan YG, Lin JJ, Su JH, Wu WT, Wu YJ. Suppression of Migration and Invasion by 4-Carbomethoxyl-10-Epigyrosanoldie E from the Cultured Soft Coral Sinularia sandensis through the MAPKs Pathway on Oral Cancer Cells. Adv Pharmacol Pharm Sci 2024; 2024:6695837. [PMID: 38374934 PMCID: PMC10876307 DOI: 10.1155/2024/6695837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/27/2023] [Accepted: 01/20/2024] [Indexed: 02/21/2024] Open
Abstract
The primary reason for cancer-related fatalities is metastasis. The compound 4-carbomethoxyl-10-epigyrosanoldie E, derived from the Sinularia sandensis soft coral species grown in cultures, exhibits properties that counteract inflammation. Moreover, it has been observed to trigger both apoptosis and autophagy within cancerous cells. This research focuses on examining the inhibitory impact of 4-carbomethoxyl-10-epigyrosanoldie E on the migration and invasion processes in Cal-27 and Ca9-22 oral cancer cell lines. To assess how this compound affects cell migration and invasion, the Boyden chamber assay was employed. Furthermore, Western blot analysis was utilized to explore the underlying molecular mechanisms. In a dose-dependent manner, 4-carbomethoxyl-10-epigyrosanoldie E notably decreased the levels of matrix metalloproteinase-2 (MMP-2) and MMP-9, along with urokinase-type plasminogen activator (uPA), in both Cal-27 and Ca9-22 cell lines. Conversely, it elevated the concentrations of tissue inhibitors of metalloproteinases-1 (TIMP-1) and TIMP-2. In addition, the treatment with this compound led to the inhibition of phosphorylation in extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). It also curtailed the expression of several key proteins including focal adhesion kinase (FAK), protein kinase C (PKC), growth factor receptor-bound protein 2 (GRB2), Rac, Ras, Rho A, mitogen-activated protein kinase kinase kinase 3 (MEKK3), and mitogen-activated protein kinase kinase 7 (MKK7). Furthermore, the expression levels of IQ-domain GTPase-activating protein 1 (IQGAP1) and zonula occludens-1 (ZO-1) were significantly reduced by the compound. The ability of 4-carbomethoxyl-10-epigyrosanoldie E to inhibit the migration and invasion of Cal-27 and Ca9-22 oral cancer cells was observed to be dose dependent. This inhibitory effect is primarily attributed to the suppression of MMP-2 and MMP-9 expression, as well as the downregulation of the mitogen-activated protein kinase (MAPK) signaling pathway.
Collapse
Affiliation(s)
- Rou-Yi Fang
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Pingtung Branch, Kaohsiung, Taiwan
- Department of Nursing, Meiho University, Pingtung, Taiwan
| | - Yueh-Wen Liu
- Department of Cosmetics and Fashion Styling, Cheng Shiu University, Kaohsiung, Taiwan
| | - Yih-Gang Goan
- Division of Thoracic Surgery Department of Surgery, Pingtung Veterans General Hospital, Pingtung, Taiwan
| | - Jen-Jie Lin
- Yu Jun Biotechnology Co., Ltd., Kaohsiung, Taiwan
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan
| | - Wen-Tung Wu
- Department of Food Science and Nutrition, Meiho University, Pingtung 91202, Taiwan
| | - Yu-Jen Wu
- Yu Jun Biotechnology Co., Ltd., Kaohsiung, Taiwan
| |
Collapse
|
7
|
Oh SY, Na SB, Kang YK, Do JT. In Vitro Embryogenesis and Gastrulation Using Stem Cells in Mice and Humans. Int J Mol Sci 2023; 24:13655. [PMID: 37686459 PMCID: PMC10563085 DOI: 10.3390/ijms241713655] [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: 08/01/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
During early mammalian embryonic development, fertilized one-cell embryos develop into pre-implantation blastocysts and subsequently establish three germ layers through gastrulation during post-implantation development. In recent years, stem cells have emerged as a powerful tool to study embryogenesis and gastrulation without the need for eggs, allowing for the generation of embryo-like structures known as synthetic embryos or embryoids. These in vitro models closely resemble early embryos in terms of morphology and gene expression and provide a faithful recapitulation of early pre- and post-implantation embryonic development. Synthetic embryos can be generated through a combinatorial culture of three blastocyst-derived stem cell types, such as embryonic stem cells, trophoblast stem cells, and extraembryonic endoderm cells, or totipotent-like stem cells alone. This review provides an overview of the progress and various approaches in studying in vitro embryogenesis and gastrulation in mice and humans using stem cells. Furthermore, recent findings and breakthroughs in synthetic embryos and gastruloids are outlined. Despite ethical considerations, synthetic embryo models hold promise for understanding mammalian (including humans) embryonic development and have potential implications for regenerative medicine and developmental research.
Collapse
Affiliation(s)
| | | | | | - Jeong Tae Do
- Department of Stem Cell Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Republic of Korea; (S.Y.O.); (S.B.N.); (Y.K.K.)
| |
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: 11] [Impact Index Per Article: 5.5] [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
|
Lian X, Zhang W, He-Yang J, Zhou X. Human milk oligosaccharide disialyllacto-n-tetraose protects human intestinal epithelium integrity and permeability against mast cell chymase-induced disruption by stabilizing ZO-1/FAK/P38 pathway of intestinal epithelial cell. Immunopharmacol Immunotoxicol 2022:1-10. [PMID: 36537314 DOI: 10.1080/08923973.2022.2160730] [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: 12/24/2022]
Abstract
CONTEXT Inflammatory bowel disease (IBD) is a chronic gut disease with intestinal-epithelium disruption. Mast cell (MC) has been discussed in IBD studies, but its subset MCTC (chymase/tryptase) and MC-chymase have not been well-explored extensively. Human-milk-oligosaccharide-Disialyllacto-N-Tetraose (DSLNT) was reported as an effective strategy to protect infants against IBD with unclear mechanism. OBJECTIVE This study was to examine the distribution of chymase-positive mast cells in the intestinal-epithelium-tissue of IBD infants, to explore the MC-chymase function on intestinal-epithelium, and to investigate the influences of DSLNT against MC-chymase-induced disruptions. MATERIALS AND METHODS The intestinal-biopsies (surgical-waste) of the infants with IBD or with intestinal-atresia (non-IBD) were paraffin-embedded for immunohistochemistry. In-situ intestinal-tissue model and in-vitro human-intestinal-epithelial-cell (Caco-2) model were established with or without the treatments of MC-chymase (50mU/mL), DSLNT (600 µM) and DSLNT + MC-chymase respectively. The tissue morphology analysis, cell proliferation assay, cell-gap-closure assessment, fluorescence-immunocytochemistry, western blot, trans-epithelial-electrical-resistance, cell-cycle and statistical analysis were applied. RESULTS There was an increased number of MCTC subset around the inflamed intestinal area in-vivo; MC-chymase caused intestinal-epithelial-barrier damage in-situ, decreased trans-epithelial-electrical-resistance of caco-2 cell monolayer in-vitro; while DSLNT protected epithelium against MC-chymase induced disruptions. MC-chymase reduced cell-viability, proliferation and migration, altered cell-cycle, down-regulated ZO-1, FAK, and P38 expressions, while DSLNT protected cells by impairing MC-chymase-induced interruptions. DSLNT can rescue ZO-1, FAK and P38 expressions and restore epithelial-cell integrity and cell cycle. CONCLUSIONS Chymase-positive MCs are involved in IBD progress. MC-chymase disrupts intracellular ZO-1/FAK/P38 signal pathway and cell-cell/cell-matrix contacts, while DSLNT protects intestinal-epithelium against MC-chymase to maintain the intestinal epithelium integrity.
Collapse
Affiliation(s)
- Xuejiao Lian
- The School of Pharmacy, Changzhou University, Jiangsu, China
| | - Wenting Zhang
- The School of Pharmacy, Changzhou University, Jiangsu, China.,Department of Pharmacy, Changzhou Children's Hospital, Changzhou, China
| | - Jingqiu He-Yang
- The School of Pharmacy, Changzhou University, Jiangsu, China
| | - Xiaoying Zhou
- The School of Pharmacy, Changzhou University, Jiangsu, China
| |
Collapse
|
10
|
Direct Injection of Recombinant AAV-Containing Solution into the Oviductal Lumen of Pregnant Mice Caused In Situ Infection of Both Preimplantation Embryos and Oviductal Epithelium. Int J Mol Sci 2022; 23:ijms23094897. [PMID: 35563284 PMCID: PMC9105285 DOI: 10.3390/ijms23094897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Adeno-associated virus (AAV) vector is an efficient viral-based gene delivery tool used with many types of cells and tissues, including neuronal cells and muscles. AAV serotype 6 (AAV-6), one of numerous AAV serotypes, was recently found to efficiently transduce mouse preimplantation embryos. Furthermore, through coupling with a clustered, regularly interspaced, short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) system—a modern genome editing technology—AAV-6 has been shown to effectively create a mutation at a target locus, which relies on isolation of zygotes, in vitro viral infection, and transplantation of the infected embryos to recipient females. Unfortunately, this procedure, termed “ex vivo handling of embryos”, requires considerable investment of capital, time, and effort. Direct transduction of preimplantation embryos through the introduction of AAV-6 into the oviductal lumen of pregnant females would be an ideal approach. In this study, we injected various types of recombinant AAV vectors (namely, rAAV-CAG-EGFP-1, -2, -5, and -6, each carrying an enhanced green fluorescent protein [EGFP] cDNA whose expression is under the influence of a cytomegalovirus enhancer + chicken β-actin promoter) into the ampulla region of oviducts in pregnant female mice at Day 0.7 of pregnancy (corresponding to the late 1-cell stage), and EGFP-derived green fluorescence was assessed in the respective morulae. The highest levels of fluorescence were observed in rAAV-CAG-EGFP-6. The oviductal epithelium was distinctly fluorescent. The fluorescence in embryos peaked at the morula stage. Our results indicate that intra-oviductal injection of AAV-6 vectors is the most effective method for transducing zona pellucida-enclosed preimplantation embryos in situ. AAV-6 vectors could be a useful tool in the genetic manipulation of early embryos, as well as oviductal epithelial cells.
Collapse
|
11
|
Calder MD, Chen R, MacDonald A, MacNeily Z, Leung ZCL, Adus S, Cui S, Betts DH, Rafea BA, Watson AJ. Effects of palmitic acid on localization of embryo cell fate and blastocyst formation gene products. Reproduction 2022; 163:133-143. [PMID: 35038315 PMCID: PMC8859922 DOI: 10.1530/rep-21-0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/13/2022] [Indexed: 12/04/2022]
Abstract
As obese and overweight patients commonly display hyperlipidemia and are increasingly accessing fertility clinics for their conception needs, our studies are directed at understanding the effects of hyperlipidemia on early pregnancy. We have focused on investigating palmitic acid (PA) and oleic acid (OA) treatment alone and in combination from the mouse two-cell stage embryos as a model for understanding their effects on the mammalian preimplantation embryo. We recently reported that PA exerts a negative effect on mouse two-cell progression to the blastocyst stage, whereas OA co-treatment reverses that negative effect. In the present study, we hypothesized that PA treatment of mouse embryos would disrupt proper localization of cell fate determining and blastocyst formation gene products and that co-treatment with OA would reverse these effects. Our results demonstrate that PA treatment significantly (P < 0.05) reduces blastocyst development and cell number but did not prevent nuclear localization of YAP in outer cells. PA treatment significantly reduced the number of OCT4+ and CDX2+ nuclei. PA-treated embryos had lower expression of blastocyst formation proteins (E-cadherin, ZO-1 and Na/K-ATPase alpha1 subunit). Importantly, co-treatment of embryos with OA reversed PA-induced effects on blastocyst development and increased inner cell mass (ICM) and trophectoderm (TE) cell numbers and expression of blastocyst formation proteins. Our findings demonstrate that PA treatment does not impede cell fate gene localization but does disrupt proper blastocyst formation gene localization during mouse preimplantation development. OA treatment is protective and reverses PA's detrimental effects. The results advance our understanding of the impact of FFA exposure on mammalian preimplantation development.
Collapse
Affiliation(s)
- Michele D Calder
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Robert Chen
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Anastasia MacDonald
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Zoe MacNeily
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Zuleika Chin Lai Leung
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Samira Adus
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Shiyu Cui
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Dean H Betts
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Basim Abu Rafea
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew J Watson
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- The Children’s Health Research Institute – Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
12
|
Yang M, Yu H, Yu X, Liang S, Hu Y, Luo Y, Izsvák Z, Sun C, Wang J. Chemical-induced chromatin remodeling reprograms mouse ESCs to totipotent-like stem cells. Cell Stem Cell 2022; 29:400-418.e13. [PMID: 35143761 DOI: 10.1016/j.stem.2022.01.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 10/18/2021] [Accepted: 01/18/2022] [Indexed: 11/03/2022]
Abstract
Totipotent cells have more robust developmental potency than any other cell types, giving rise to both embryonic and extraembryonic tissues. Stable totipotent cell cultures and deciphering the principles of totipotency regulation would be invaluable to understand cell plasticity and lineage segregation in early development. Our approach of remodeling the pericentromeric heterochromatin and re-establishing the totipotency-specific broad H3K4me3 domains promotes the pluri-to-totipotency transition. Our protocol establishes a closer match of mouse 2-cell (2C) embryos than any other 2C-like cells. These totipotent-like stem cells (TLSCs) are stable in culture and possess unique molecular features of the mouse 2C embryo. Functionally, TLSCs are competent for germline transmission and give rise to both embryonic and extraembryonic lineages at high frequency. Therefore, TLSCs represent a highly valuable cell type for studies of totipotency and embryology.
Collapse
Affiliation(s)
- Mingzhu Yang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hanwen Yu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiu Yu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shiqi Liang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanlang Hu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuxin Luo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13092, Germany
| | - Chuanbo Sun
- Guangzhou Institute of Pediatrics, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510623, China.
| | - Jichang Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
13
|
Cell fate determination and Hippo signaling pathway in preimplantation mouse embryo. Cell Tissue Res 2021; 386:423-444. [PMID: 34586506 DOI: 10.1007/s00441-021-03530-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
First cell fate determination plays crucial roles in cell specification during early phases of embryonic development. Three classical concepts have been proposed to explain the lineage specification mechanism of the preimplantation embryo: inside-outside, pre-patterning, and polarity models. Transcriptional effectors of the Hippo signal pathway are YAP and TAZ activators that can create a shuttle between the cytoplasm and the nucleus. Despite different localizations of YAP in the cell, it determines the fate of ICM and TE. How the decisive cue driving factors that determine YAP localization are coordinated remains a central unanswered question. How can an embryonic cell find its position? The objective of this review is to summarize the molecular and mechanical aspects in cell fate decision during mouse preimplantation embryonic development. The findings will reveal the relationship between cell-cell adhesion, cell polarity, and determination of cell fate during early embryonic development in mice and elucidate the inducing/inhibiting mechanisms that are involved in cell specification following zygotic genome activation and compaction processes. With future studies, new biophysical and chemical cues in the cell fate determination will impart significant spatiotemporal effects on early embryonic development. The achieved knowledge will provide important information to the development of new approaches to be used in infertility treatment and increase the success of pregnancy.
Collapse
|
14
|
Abstract
Extended/expanded pluripotent stem (EPS) cells can efficiently contribute to both embryonic and extraembryonic lineages in vitro and in vivo. Starting from these cells, we established a 3D differentiation system that enabled the generation of blastocyst-like structures (EPS-blastoids) through lineage segregation and self-organization. We also provide proof of concept that EPS-blastoids can be generated from adult cells via cellular reprogramming. EPS-blastoids provide a unique platform for studying early embryogenesis. For complete details on the use and execution of this protocol, please refer to Li et al. (2019). A protocol that enables the generation of blastocyst-like structures from EPS cells EPS cell aggregates recapitulate early developmental events in vitro to form blastoids EPS-blastoids resemble blastocysts in morphology and cell lineage allocation EPS-blastoids are able to implant in utero
Collapse
|
15
|
Abstract
The cytoskeleton - comprising actin filaments, microtubules and intermediate filaments - serves instructive roles in regulating cell function and behaviour during development. However, a key challenge in cell and developmental biology is to dissect how these different structures function and interact in vivo to build complex tissues, with the ultimate aim to understand these processes in a mammalian organism. The preimplantation mouse embryo has emerged as a primary model system for tackling this challenge. Not only does the mouse embryo share many morphological similarities with the human embryo during its initial stages of life, it also permits the combination of genetic manipulations with live-imaging approaches to study cytoskeletal dynamics directly within an intact embryonic system. These advantages have led to the discovery of novel cytoskeletal structures and mechanisms controlling lineage specification, cell-cell communication and the establishment of the first forms of tissue architecture during development. Here we highlight the diverse organization and functions of each of the three cytoskeletal filaments during the key events that shape the early mammalian embryo, and discuss how they work together to perform key developmental tasks, including cell fate specification and morphogenesis of the blastocyst. Collectively, these findings are unveiling a new picture of how cells in the early embryo dynamically remodel their cytoskeleton with unique spatial and temporal precision to drive developmental processes in the rapidly changing in vivo environment.
Collapse
|
16
|
Gertsenstein M, Nutter LMJ. Production of knockout mouse lines with Cas9. Methods 2021; 191:32-43. [PMID: 33524495 DOI: 10.1016/j.ymeth.2021.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/05/2020] [Accepted: 01/13/2021] [Indexed: 12/26/2022] Open
Abstract
Knockout mice are used extensively to explore the phenotypic effects of mammalian gene dysfunction. With the application of RNA-guided Cas9 nuclease technology for the production of knockout mouse lines, the time, as well as the resources needed, to progress from identification of a gene of interest to production of a knockout line is significantly reduced. Here we present our standard methodology to produce knockout mouse lines by the electroporation of Cas9 ribonucleoprotein (RNP) into mouse zygotes. Using this protocol, we have obtained an 80% success rate in the generation of founders for null alleles with a subsequent 93% germline transmission rate. These methods rely on equipment already present in the majority of transgenic facilities and should be straightforward to implement where appropriate embryo handling expertise exists.
Collapse
Affiliation(s)
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto M5T 3H7, Canada; The Hospital for Sick Children, Toronto M5G 1X8, Canada
| |
Collapse
|
17
|
Van Heertum K, Lam L, Richardson B, Cartwright MJ, Mesiano SA, Cameron MJ, Weinerman R. Blastocyst Vitrification and Trophectoderm Biopsy Cumulatively Alter Embryonic Gene Expression in a Mouse Model. Reprod Sci 2021; 28:2961-2971. [PMID: 33826099 DOI: 10.1007/s43032-021-00560-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/25/2021] [Indexed: 11/25/2022]
Abstract
Although embryo vitrification has been used extensively in human assisted reproductive technology (ART) and animal models, epidemiologic evidence and randomized controlled trials suggest differences in pregnancy/perinatal outcomes (birthweight, risk for preterm birth, and pre-eclampsia) between babies born from fresh versus frozen embryo transfers. To address the uncertainty surrounding the effects of laboratory manipulations of embryos on clinical outcomes, we subjected mouse blastocysts to increasing levels of manipulation for transcriptome analysis. Blastocysts were randomly divided into four groups: no manipulation (control), single vitrification/thaw (1 vit), double vitrification/thaw (2 vit), and single vitrification/thaw plus trophectoderm biopsy and again vitrified/thawed (2 vit + bx). Three sets of 15 blastocysts in each group were pooled for RNA sequencing, and differentially expressed genes (DEGs) and pathways were determined by statistical analysis. Blastocysts were also stained for ZO-1 and F-actin to assess cytoskeletal integrity. Freeze/thaw and biopsy manipulation affected multiple biological pathways. The most significant differences were detected in genes related to innate immunity, apoptosis, and mitochondrial function, with the magnitude of change proportional to the extent to manipulation. Significant disruptions were also seen in cytoskeletal staining, with greater disruptions seen with greater of manipulation. Our data suggests that embryo vitrification and biopsy affect embryo gene transcription, with several identified DEGs that may have plausible mechanisms for the clinical outcomes seen in human offspring following ART. Further study is required to determine whether these alterations in gene expression are associated with clinical differences seen in children born from fresh or frozen embryo transfer.
Collapse
Affiliation(s)
- Kristin Van Heertum
- University Hospitals Fertility Center/Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lisa Lam
- CCRM New York Fertility, New York, NY, 10019, USA
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michael J Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sam A Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rachel Weinerman
- University Hospitals Fertility Center/Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
18
|
Hakim BA, Tyagi V, Agnihotri SK, Nath A, Agrawal AK, Jain A, Singh D, Konwar R, Sachdev M. Electroporation of Mouse Follicles, Oocytes and Embryos without Manipulating Zona Pellucida. J Dev Biol 2021; 9:jdb9020013. [PMID: 33915920 PMCID: PMC8167572 DOI: 10.3390/jdb9020013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
Electroporation is an effective technique of transfection, but its efficiency depends on the optimization of various parameters. In this study, a simplified and efficient method of gene manipulation was standardized through electroporation to introduce a recombinant green fluorescent protein (GFP) construct as well as RNA-inhibitors in intact mouse follicles, oocytes and early embryos, where various electroporation parameters like voltage, pulse number and pulse duration were standardized. Electroporated preantral follicles were cultured further in vitro to obtain mature oocytes and their viability was confirmed through the localization of a known oocyte maturation marker, ovastacin, which appeared to be similar to the in vivo-derived mature oocytes and thus proved the viability of the in vitro matured oocytes after electroporation. Standardized electroporation parameters, i.e., three pulses of 30 V for 1 millisecond at an interval of 10 s, were applied to manipulate the expression of mmu-miR-26a in preantral follicles through the electroporation of miR inhibitors and mimics. The TUNEL apoptosis assay confirmed the normal development of the electroporated embryos when compared to the normal embryos. Conclusively, for the first time, this study demonstrated the delivery of exogenous oligonucleotides into intact mouse follicles, oocytes and embryos without hampering their zona pellucida (ZP) and further development.
Collapse
Affiliation(s)
- Bilal Ahmad Hakim
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Vaishali Tyagi
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Saurabh Kumar Agnihotri
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Amar Nath
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Ankit Kumar Agrawal
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Ankita Jain
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Deependra Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Rituraj Konwar
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Monika Sachdev
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
- Correspondence: ; Tel.: +91-9026561774
| |
Collapse
|
19
|
Cao L, Yang T, Huang S, Yun X, Hou H, Wang T, Shi D, Li X. Expression patterns of ZO-1/2 and their effects on porcine oocyte in vitro maturation and early embryonic development. Theriogenology 2020; 161:262-270. [PMID: 33348145 DOI: 10.1016/j.theriogenology.2020.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/30/2022]
Abstract
Zonula occludens (ZO)-1 and ZO-2 are involved in epithelial polarity maintenance, gene transcription, cell proliferation and tumor cell metastasis. Regulating ZO-1/2 expression influences the early embryonic development of mice, but whether they are involved in oocyte maturation is still poorly understood. In the present study, the expression patterns of ZO-1 and ZO-2 in porcine cumulus cells and oocytes matured in vitro and early embryos from parthenogenetic activation were detected by qRT-PCR or Western blot, and then their roles in porcine oocyte maturation and early embryo development were investigated by shRNA technology. ZO-1 and ZO-2 were found to be expressed in cumulus cells, oocytes and early embryos, while ZO-1α+ was expressed only in cumulus cells, morula and blastocysts. During in vitro maturation (IVM), the abundance of ZO-1 and ZO-2 in oocytes was significantly higher than that in cumulus cells at 0 h (P < 0.01), and their mRNA and protein levels displayed relatively higher expression at 0 and 18 h, respectively. Compared with the control groups, cumulus cell expansion, oocyte nucleus maturation, and subsequent cleavage were not influenced by treatment of the cumulus-oocyte complexes (COCs) with ZO-1-shRNA1, ZO-2-shRNA2 or combined ZO-1-shRNA1 and ZO-2-shRNA2 lentivirus (P > 0.05). However, the blastocyst rate was reduced by treatment of COCs with ZO-1-shRNA1 but not ZO-2-shRNA2. The total cell number of blastocysts was decreased by downregulation of ZO-1 and ZO-2 (P < 0.05). Downregulation of ZO-1 and ZO-2 also resulted in a significant decrease (P < 0.05) in the expression of Cx43, Cx45, PTX3 and PTGS2 in cumulus cells, Cx45, BMP15, ZP3 and C-KIT in MII oocytes, and Nanog in blastocysts, with the exception of HAS2 expression in cumulus cells and Oct4 expression in blastocysts (P > 0.05). Altogether, the above results indicate that ZO-1 and ZO-2 display similar expression patterns during porcine oocyte IVM and are critical to porcine oocyte maturation and early embryonic development.
Collapse
Affiliation(s)
- Lihua Cao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China
| | - Ting Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China
| | - Shihai Huang
- College of Life Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Xuedan Yun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China
| | - Hanqi Hou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China
| | - Ting Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China.
| | - Xiangping Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, 530005, PR China.
| |
Collapse
|
20
|
Tocci A. The unknown human trophectoderm: implication for biopsy at the blastocyst stage. J Assist Reprod Genet 2020; 37:2699-2711. [PMID: 32892265 DOI: 10.1007/s10815-020-01925-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/11/2020] [Indexed: 11/30/2022] Open
Abstract
Trophectoderm biopsy is increasingly performed for pre-implantation genetic testing of aneuploidies and considered a safe procedure on short-term clinical outcome, without strong assessment of long-term consequences. Poor biological information on human trophectoderm is available due to ethical restrictions. Therefore, most studies have been conducted in vitro (choriocarcinoma cell lines, embryonic and pluripotent stem cells) and on murine models that nevertheless poorly reflect the human counterpart. Polarization, compaction, and blastomere differentiation (e.g., the basis to ascertain trophectoderm origin) are poorly known in humans. In addition, the trophectoderm function is poorly known from a biological point of view, although a panoply of questionable and controversial microarray studies suggest that important genes overexpressed in trophectoderm are involved in pluripotency, metabolism, cell cycle, endocrine function, and implantation. The intercellular communication system between the trophectoderm cells and the inner cell mass, modulated by cell junctions and filopodia in the murine model, is obscure in humans. For the purpose of this paper, data mainly on primary cells from human and murine embryos has been reviewed. This review suggests that the trophectoderm origin and functions have been insufficiently ascertained in humans so far. Therefore, trophectoderm biopsy should be considered an experimental procedure to be undertaken only under approved rigorous experimental protocols in academic contexts.
Collapse
Affiliation(s)
- Angelo Tocci
- Reproductive Medicine Unit, Gruppo Donnamed, Via Giuseppe Silla 12, Rome, Italy.
| |
Collapse
|
21
|
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: 27] [Impact Index Per Article: 5.4] [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
|
22
|
Özgüç Ö, Maître JL. Multiscale morphogenesis of the mouse blastocyst by actomyosin contractility. Curr Opin Cell Biol 2020; 66:123-129. [PMID: 32711300 DOI: 10.1016/j.ceb.2020.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/20/2020] [Accepted: 05/05/2020] [Indexed: 01/31/2023]
Abstract
During preimplantation development, the mouse embryo forms the blastocyst, which consists of a squamous epithelium enveloping a fluid-filled lumen and a cluster of pluripotent cells. The shaping of the blastocyst into its specific architecture is a prerequisite to implantation and further development of the embryo. Recent studies identified the central role of the actomyosin cortex in generating the forces driving the successive steps of blastocyst morphogenesis. As seen in other developing animals, actomyosin functions across spatial scales from the subcellular to the tissue levels. In addition, the slow development of the mouse embryo reveals that actomyosin contractility operates at multiple timescales with periodic cortical waves of contraction every ∼80 s and tissue remodeling over hours.
Collapse
Affiliation(s)
- Özge Özgüç
- Institut Curie, 26, rue d'Ulm - 75248 Paris Cedex 05 - France
| | | |
Collapse
|
23
|
Li R, Zhong C, Yu Y, Liu H, Sakurai M, Yu L, Min Z, Shi L, Wei Y, Takahashi Y, Liao HK, Qiao J, Deng H, Nuñez-Delicado E, Rodriguez Esteban C, Wu J, Izpisua Belmonte JC. Generation of Blastocyst-like Structures from Mouse Embryonic and Adult Cell Cultures. Cell 2020; 179:687-702.e18. [PMID: 31626770 DOI: 10.1016/j.cell.2019.09.029] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 06/19/2019] [Accepted: 09/21/2019] [Indexed: 11/19/2022]
Abstract
A single mouse blastomere from an embryo until the 8-cell stage can generate an entire blastocyst. Whether laboratory-cultured cells retain a similar generative capacity remains unknown. Starting from a single stem cell type, extended pluripotent stem (EPS) cells, we established a 3D differentiation system that enabled the generation of blastocyst-like structures (EPS-blastoids) through lineage segregation and self-organization. EPS-blastoids resembled blastocysts in morphology and cell-lineage allocation and recapitulated key morphogenetic events during preimplantation and early postimplantation development in vitro. Upon transfer, some EPS-blastoids underwent implantation, induced decidualization, and generated live, albeit disorganized, tissues in utero. Single-cell and bulk RNA-sequencing analysis revealed that EPS-blastoids contained all three blastocyst cell lineages and shared transcriptional similarity with natural blastocysts. We also provide proof of concept that EPS-blastoids can be generated from adult cells via cellular reprogramming. EPS-blastoids provide a unique platform for studying early embryogenesis and pave the way to creating viable synthetic embryos by using cultured cells.
Collapse
Affiliation(s)
- Ronghui Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Cuiqing Zhong
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yang Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Haisong Liu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Masahiro Sakurai
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Leqian Yu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zheying Min
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Lei Shi
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Universidad Catolica, San Antonio de Murcia, Campus de los Jeronimos, 135, Guadalupe 30107, Murcia, Spain
| | - Yulei Wei
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Yuta Takahashi
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Hsin-Kai Liao
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Universidad Catolica, San Antonio de Murcia, Campus de los Jeronimos, 135, Guadalupe 30107, Murcia, Spain
| | - Jie Qiao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Hongkui Deng
- University of Peking, 5 Yiheyuan Rd, Haidian Qu, Beijing 100871, China
| | - Estrella Nuñez-Delicado
- Universidad Catolica, San Antonio de Murcia, Campus de los Jeronimos, 135, Guadalupe 30107, Murcia, Spain
| | | | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | |
Collapse
|
24
|
Maternal Yes-Associated Protein Participates in Porcine Blastocyst Development via Modulation of Trophectoderm Epithelium Barrier Function. Cells 2019; 8:cells8121606. [PMID: 31835702 PMCID: PMC6952962 DOI: 10.3390/cells8121606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023] Open
Abstract
The establishment of a functional trophectoderm (TE) epithelium is an essential prerequisite for blastocyst formation and placentation. Transcription coactivator yes-associated protein (YAP), a downstream effector of the hippo signaling pathway, is required for specification of both the TE and epiblast lineages in mice. However, the biological role of YAP in porcine blastocyst development is not known. Here, we report that maternally derived YAP protein is localized to both the cytoplasm and nuclei prior to the morula stage and is then predominantly localized to the TE nuclei in blastocysts. Functionally, maternal YAP knockdown severely impeded blastocyst formation and perturbed the allocation of the first two lineages. The treatment of embryos with verteporfin, a pharmacological inhibitor of YAP, faithfully recapitulated the phenotype observed in YAP deleted embryos. Mechanistically, we found that maternal YAP regulates multiple genes which are important for lineage commitment, tight junction assembly, and fluid accumulation. Consistent with the effects on tight junction gene expression, a permeability assay revealed that paracellular sealing was defective in the trophectoderm epithelium. Lastly, YAP knockdown in a single blastomere at the 2-cell stage revealed that the cellular progeny of the YAP+ blastomere were sufficient to sustain blastocyst formation via direct complementation of the defective trophectoderm epithelium. In summary, these findings demonstrate that maternal YAP facilitates porcine blastocyst development through transcriptional regulation of key genes that are essential for lineage commitment, tight junction assembly, and fluid accumulation.
Collapse
|
25
|
Guo S, Liu S, Bou G, Guo J, Jiang L, Chai Z, Cai M, Mu Y, Liu Z. Fetal bovine serum promotes the development of in vitro porcine blastocysts by activating the Rho-associated kinase signalling pathway. Reprod Fertil Dev 2019; 31:366-376. [PMID: 30253120 DOI: 10.1071/rd18070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/13/2018] [Indexed: 02/01/2023] Open
Abstract
Fetal bovine serum (FBS) supplementation has beneficial effects on invitro porcine embryonic development, but the underlying mechanisms are unclear. In the present study we found that the addition of FBS to PZM-3 increased the number of cells in porcine blastocysts and hatching rate invitro primarily by promoting proliferation of the inner cell mass and further differentiation. Moreover, based on the following results, we surmise that FBS benefits blastocyst development by activating Rho-associated kinase (ROCK) signalling: (1) the ROCK signalling inhibitor Y-27632 decreased the blastocyst rate and the number of cells in blastocysts, whereas FBS rescued the developmental failure induced by Y-27632; (2) the mRNA levels of two ROCK isoforms, ROCK1 and ROCK2, were significantly increased in blastocysts derived from medium containing FBS; and (3) FBS increased RhoA/Rho-kinase expression in the nucleus of embryonic cells. These results indicate that FBS promotes the invitro development of porcine embryos by activating ROCK signalling in a chemically defined medium.
Collapse
Affiliation(s)
- Shimeng Guo
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Shichao Liu
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Gerelchimeg Bou
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Jia Guo
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Liyuan Jiang
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Zhuang Chai
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Mingming Cai
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Yanshuang Mu
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China
| |
Collapse
|
26
|
Jeong Y, Ock S, Yoo JG, Yu D, Choi I. The Cxadr–Adam10 complex plays pivotal roles in tight junction integrity and early trophoblast development in mice. Mol Reprod Dev 2019; 86:1628-1638. [DOI: 10.1002/mrd.23250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 07/16/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Yelin Jeong
- Division of Animal and Dairy Sciences, College of Agriculture and Life SciencesChungnam National UniversityDaejeon Republic of Korea
- Disease Model Research Laboratory, Genome Editing Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon Republic of Korea
| | - Sun‐A Ock
- National Institute of Animal ScienceRural Development AdministrationJeollabuk‐do Republic of Korea
| | - Jae Gyu Yoo
- National Institute of Animal ScienceRural Development AdministrationJeollabuk‐do Republic of Korea
| | - Dae‐Yeul Yu
- Disease Model Research Laboratory, Genome Editing Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon Republic of Korea
- Department of Functional GenomicsUniversity of Science and TechnologyDaejeon Republic of Korea
| | - Inchul Choi
- Division of Animal and Dairy Sciences, College of Agriculture and Life SciencesChungnam National UniversityDaejeon Republic of Korea
| |
Collapse
|
27
|
Kwon J, Park S, Seong MJ, Choi I, Kim NH. Cytoplasmic polyadenylation element binding protein 2 (CPEB2) is required for tight-junction assembly for establishment of porcine trophectoderm epithelium. Reprod Fertil Dev 2019; 31:412-419. [DOI: 10.1071/rd18098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/27/2018] [Indexed: 11/23/2022] Open
Abstract
Cytoplasmic polyadenylation element binding protein (CPEB) is an RNA-binding protein that promotes elongation of poly(A) tails and regulates mRNA translation. CPEB depletion in mammary epithelium is known to disrupt tight-junction (TJ) assembly via mislocalisation of tight junction protein 1 (TJP1), but the role of CPEB in the biological functions associated with TJs has not yet been studied. The objective of this study was to investigate the roles of CPEB2 during porcine parthenote development. CPEB2 was detected in both the nuclei and apical cytoplasm at the 4- and 8-cell stages and was localised to cell–cell contact after the initiation of the morula stage. Its depletion led to retarded blastocyst formation caused by impaired TJ assembly. Moreover, transcription of TJ-associated genes, including TJP1, Coxsackie virus and adenovirus receptor (CXADR) and occludin (OCLN), was not affected, but the corresponding proteins were not properly localised at the apical cell membrane in morulae, suggesting that CPEB2 confers mRNA stability or determines subcellular localisation for translation. Remarkably reduced relative levels of TJP1 transcripts bearing the 3′-untranslated region were noted, indicating that CPEB2 mediates TJP1 mRNA stability. In conclusion, our findings demonstrate that because of its regulation of TJP1, CPEB2 is required for TJ assembly during porcine blastocyst development.
Collapse
|
28
|
White MD, Zenker J, Bissiere S, Plachta N. Instructions for Assembling the Early Mammalian Embryo. Dev Cell 2018; 45:667-679. [DOI: 10.1016/j.devcel.2018.05.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/23/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022]
|
29
|
Maître JL. Mechanics of blastocyst morphogenesis. Biol Cell 2017; 109:323-338. [DOI: 10.1111/boc.201700029] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Jean-Léon Maître
- Institut Curie; PSL Research University; CNRS UMR3215, INSERM U934; Paris France
| |
Collapse
|
30
|
Peng H, Liu H, Liu F, Gao Y, Chen J, Huo J, Han J, Xiao T, Zhang W. NLRP2 and FAF1 deficiency blocks early embryogenesis in the mouse. Reproduction 2017. [PMID: 28630100 DOI: 10.1530/rep-16-0629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nlrp2 is a maternal effect gene specifically expressed by mouse ovaries; deletion of this gene from zygotes is known to result in early embryonic arrest. In the present study, we identified FAF1 protein as a specific binding partner of the NLRP2 protein in both mouse oocytes and preimplantation embryos. In addition to early embryos, both Faf1 mRNA and protein were detected in multiple tissues. NLRP2 and FAF1 proteins were co-localized to both the cytoplasm and nucleus during the development of oocytes and preimplantation embryos. Co-immunoprecipitation assays were used to confirm the specific interaction between NLRP2 and FAF1 proteins. Knockdown of the Nlrp2 or Faf1 gene in zygotes interfered with the formation of a NLRP2-FAF1 complex and led to developmental arrest during early embryogenesis. We therefore conclude that NLRP2 interacts with FAF1 under normal physiological conditions and that this interaction is probably essential for the successful development of cleavage-stage mouse embryos. Our data therefore indicated a potential role for NLRP2 in regulating early embryo development in the mouse.
Collapse
Affiliation(s)
- Hui Peng
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Haijun Liu
- Tianjin Institute of Animal Science and Veterinary Medicine, Tianjin, People's Republic of China
| | - Fang Liu
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Yuyun Gao
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Jing Chen
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Jianchao Huo
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Jinglin Han
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Tianfang Xiao
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| | - Wenchang Zhang
- College of Animal Science, Fujian Agriculture and Forestry University, Fujian, Fuzhou, People's Republic of China
| |
Collapse
|
31
|
Shin H, Bang S, Kim J, Jun JH, Song H, Lim HJ. The formation of multivesicular bodies in activated blastocysts is influenced by autophagy and FGF signaling in mice. Sci Rep 2017; 7:41986. [PMID: 28155881 PMCID: PMC5290465 DOI: 10.1038/srep41986] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023] Open
Abstract
Dormant blastocysts during delayed implantation undergo autophagic activation, which is an adaptive response to prolonged survival in utero during less favorable environment. We observed that multivesicular bodies (MVBs) accumulate in the trophectoderm of dormant blastocysts upon activation for implantation. Since autophagosomes are shown to fuse with MVBs and efficient autophagic degradation requires functional MVBs, we examined if MVB formation in activated blastocysts are associated with protracted autophagic state during dormancy. We show here that autophagic activation during dormancy is one precondition for MVB formation in activated blastocysts. Furthermore, the blockade of FGF signaling with PD173074 partially interferes with MVB formation in these blastocysts, suggesting the involvement of FGFR signaling in this process. We believe that MVB formation in activated blastocysts after dormancy is a potential mechanism of clearing subcellular debris accumulated during prolonged autophagy.
Collapse
Affiliation(s)
- Hyejin Shin
- Department of Biomedical Science &Technology, Institute of Biomedical Science &Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Soyoung Bang
- Department of Biomedical Science &Technology, Institute of Biomedical Science &Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Jiyeon Kim
- Department of Biomedical Science &Technology, Institute of Biomedical Science &Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Jin Hyun Jun
- Department of Biomedical Laboratory Science, Eulji University, 553 Sanseong-daero, Seongnam, Gyeonggi-do 13135, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, CHA Bio Complex, 689 Sampyeong-dong, Seongnam, Gyeonggi-do 13884, Korea
| | - Hyunjung Jade Lim
- Department of Biomedical Science &Technology, Institute of Biomedical Science &Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.,Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| |
Collapse
|
32
|
Ma W, Huang X, Yang X, Liang X. Maternal obesity in mice not only affects fresh embryo quality but also aggravates injury due to vitrification. J Assist Reprod Genet 2016; 33:1515-1523. [PMID: 27544277 DOI: 10.1007/s10815-016-0790-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/08/2016] [Indexed: 12/19/2022] Open
Abstract
PURPOSE The aims of the present study are to identify the mechanism(s) whereby obesity impairs fresh embryos and to clarify the effects of vitrification on lipid droplet content within embryos from maternally obese mice. METHODS The diet-induced obesity mouse model was established, and the zygotes were captured and cultured to day 3. The eight-cell embryos were selected and divided into fresh and vitrified groups. The blastocysts derived from fresh embryos were used as a control. The expression profiles of endoplasmic reticulum (ER) stress genes (Atf4, Grp78, and Hsp70) and other genes (MnSOD, p53, Gadd45g, caspase-3, IGF-II, ZO-1, and E-cadherin) on day-3 fresh and post-warming eight-cell embryos from obese and control groups were determined. For day-5 fresh blastocysts and blastocysts previously vitrified on day 3, the expression profiles for all of the above genes were also determined. RESULTS For the fresh group, obesity significantly upregulated Hsp70, p53, IGF-II, and ZO-1 expression in embryos on day 3 and notably upregulated Atf4, MnSOD, Gadd45g, caspase-3, ZO-1, and E-cadherin expression in blastocysts on day 5. For vitrified ones, obesity significantly upregulated Atf4, MnSOD, and Gadd45g expression in embryos on day 3 and notably upregulated Hsp70 expression and downregulated MnSOD in day 5 blastocysts previously vitrified on day 3. CONCLUSIONS Obesity impairs fresh embryos and aggravates embryonic vitrification injury at a molecular level.
Collapse
Affiliation(s)
- Wenhong Ma
- Reproductive Centre of Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, 545000, People's Republic of China
| | - Xingfang Huang
- Center for Reproductive Medicine, Sixth Affiliated Hospital of Sun Yat-sen University, 17th Shou-gou-ling Road, Guangzhou, 510655, People's Republic of China
| | - Xing Yang
- Center for Reproductive Medicine, Sixth Affiliated Hospital of Sun Yat-sen University, 17th Shou-gou-ling Road, Guangzhou, 510655, People's Republic of China
| | - Xiaoyan Liang
- Center for Reproductive Medicine, Sixth Affiliated Hospital of Sun Yat-sen University, 17th Shou-gou-ling Road, Guangzhou, 510655, People's Republic of China.
| |
Collapse
|
33
|
Sato M, Ohtsuka M, Watanabe S, Gurumurthy CB. Nucleic acids delivery methods for genome editing in zygotes and embryos: the old, the new, and the old-new. Biol Direct 2016; 11:16. [PMID: 27037013 PMCID: PMC4815204 DOI: 10.1186/s13062-016-0115-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/15/2016] [Indexed: 12/31/2022] Open
Abstract
In the recent years, sequence-specific nucleases such as ZFNs, TALENs, and CRISPR/Cas9 have revolutionzed the fields of animal genome editing and transgenesis. However, these new techniques require microinjection to deliver nucleic acids into embryos to generate gene-modified animals. Microinjection is a delicate procedure that requires sophisticated equipment and highly trained and experienced technicians. Though over a dozen alternate approaches for nucleic acid delivery into embryos were attempted during the pre-CRISPR era, none of them became routinely used as microinjection. The addition of CRISPR/Cas9 to the genome editing toolbox has propelled the search for novel delivery approaches that can obviate the need for microinjection. Indeed, some groups have recently developed electroporation-based methods that have the potential to radically change animal transgenesis. This review provides an overview of the old and new delivery methods, and discusses various strategies that were attempted during the last three decades. In addition, several of the methods are re-evaluated with respect to their suitability to deliver genome editing components, particularly CRISPR/Cas9, to embryos.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544 Japan
| | - Masato Ohtsuka
- Division of Basic Molecular Science and Molecular Medicine, School of Medicine, Tokai University, Kanagawa, 259 1193 Japan
| | - Satoshi Watanabe
- Animal Genome Research Unit, Division of Animal Science, National Institute of Agrobiological Sciences, Ibaraki, 305-8602 Japan
| | - Channabasavaiah B. Gurumurthy
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
34
|
AKAP9, a Regulator of Microtubule Dynamics, Contributes to Blood-Testis Barrier Function. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:270-84. [PMID: 26687990 DOI: 10.1016/j.ajpath.2015.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/02/2015] [Accepted: 10/13/2015] [Indexed: 01/23/2023]
Abstract
The blood-testis barrier (BTB), formed between adjacent Sertoli cells, undergoes extensive remodeling to facilitate the transport of preleptotene spermatocytes across the barrier from the basal to apical compartments of the seminiferous tubules for further development and maturation into spermatozoa. The actin cytoskeleton serves unique structural and supporting roles in this process, but little is known about the role of microtubules and their regulators during BTB restructuring. The large isoform of the cAMP-responsive scaffold protein AKAP9 regulates microtubule dynamics and nucleation at the Golgi. We found that conditional deletion of Akap9 in mice after the initial formation of the BTB at puberty leads to infertility. Akap9 deletion results in marked alterations in the organization of microtubules in Sertoli cells and a loss of barrier integrity despite a relatively intact, albeit more apically localized F-actin and BTB tight junctional proteins. These changes are accompanied by a loss of haploid spermatids due to impeded meiosis. The barrier, however, progressively reseals in older Akap9 null mice, which correlates with a reduction in germ cell apoptosis and a greater incidence of meiosis. However, spermiogenesis remains defective, suggesting additional roles for AKAP9 in this process. Together, our data suggest that AKAP9 and, by inference, the regulation of the microtubule network are critical for BTB function and subsequent germ cell development during spermatogenesis.
Collapse
|
35
|
Eckert JJ, Velazquez MA, Fleming TP. Cell signalling during blastocyst morphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 843:1-21. [PMID: 25956293 DOI: 10.1007/978-1-4939-2480-6_1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Blastocyst morphogenesis is prepared for even before fertilisation. Information stored within parental gametes can influence both maternal and embryonic gene expression programmes after egg activation at fertilisation. A complex network of intrinsic, cell-cell mediated and extrinsic, embryo-environment signalling mechanisms operates throughout cleavage, compaction and cavitation. These signalling events not only ensure developmental progression, cell differentiation and lineage allocation to inner cell mass (embryo proper) and trophectoderm (future extraembryonic lineages) but also provide a degree of developmental plasticity ensuring survival in prevailing conditions by adaptive responses. Indeed, many cellular functions including differentiation, metabolism, gene expression and gene expression regulation are subject to plasticity with short- or long-term consequences even into adult life. The interplay between intrinsic and extrinsic signals impacting on blastocyst morphogenesis is becoming clearer. This has been best studied in the mouse which will be the focus of this chapter but translational significance to human and domestic animal embryology will be a focus in future years.
Collapse
Affiliation(s)
- Judith J Eckert
- Human Development and Health, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, UK
| | | | | |
Collapse
|
36
|
Jedrusik A. Making the first decision: lessons from the mouse. Reprod Med Biol 2015; 14:135-150. [PMID: 29259411 PMCID: PMC5715835 DOI: 10.1007/s12522-015-0206-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/31/2015] [Indexed: 01/06/2023] Open
Abstract
Pre-implantation development encompasses a period of 3-4 days over which the mammalian embryo has to make its first decision: to separate the pluripotent inner cell mass (ICM) from the extra-embryonic epithelial tissue, the trophectoderm (TE). The ICM gives rise to tissues mainly building the body of the future organism, while the TE contributes to the extra-embryonic tissues that support embryo development after implantation. This review provides an overview of the cellular and molecular mechanisms that control the critical aspects of this first decision, and highlights the role of critical events, namely zytotic genome activation, compaction, polarization, asymmetric cell divisions, formation of the blastocyst cavity and expression of key transcription factors.
Collapse
Affiliation(s)
- Agnieszka Jedrusik
- Wellcome Trust/CR UK Gurdon InstituteTennis Court RoadCB2 1QNCambridgeUK
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeDowning StreetCB2 3DYCambridgeUK
| |
Collapse
|
37
|
Biggins JS, Royer C, Watanabe T, Srinivas S. Towards understanding the roles of position and geometry on cell fate decisions during preimplantation development. Semin Cell Dev Biol 2015; 47-48:74-9. [PMID: 26349030 PMCID: PMC4683091 DOI: 10.1016/j.semcdb.2015.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 01/15/2023]
Abstract
The first lineage segregation event in mouse embryos produces two separate cell populations: inner cell mass and trophectoderm. This is understood to be brought about by cells sensing their position within the embryo and differentiating accordingly. The cellular and molecular underpinnings of this process remain under investigation and have variously been considered to be completely stochastic or alternately, subject to some predisposition set up at fertilisation or before. Here, we consider these views in light of recent publications, discuss the possible role of cell geometry and mechanical forces in this process and describe how modelling could contribute in addressing this issue.
Collapse
Affiliation(s)
- John S Biggins
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - Christophe Royer
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Tomoko Watanabe
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Shankar Srinivas
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Zhang H, Wang L, Li W, Mao Q, Wang Y, Li Q, Hua S, Zhang Y. A simple and efficient method to transfect small interference RNA into bovine SCNT embryos. Theriogenology 2014; 84:846-52. [PMID: 26194696 DOI: 10.1016/j.theriogenology.2014.12.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 12/09/2014] [Accepted: 12/15/2014] [Indexed: 01/12/2023]
Abstract
RNA interference is an important tool to study the gene function. Microinjection and electroporation are usually used to transfer DNA, small interference RNA (siRNA), morpholinos, and protein into oocytes or embryos. This study used a simple and effective method to transfect siRNA into bovine somatic cell nuclear transfer (SCNT) embryos. In this method, siRNA transfection and electrofusion of SCNT were combined. A pair of platinum microelectrodes was used during SCNT to complete electrofusion. A CY3-labeled siRNA-targeted DNA methyltransferase-1 (DNMT1) was chosen to verify the siRNA transfection efficiency of this approach. First, a suitable concentration of siRNA was mixed with Zimmermann's fusion medium. Reconstructed embryos were then added into the microdrops of the mixed fusion medium to simultaneously transfect the siRNA and electrofuse the SCNT embryos. Our results showed that transfecting DNMT1 siRNA via the proposed method caused obvious CY3 fluorescence and significant downregulation of DNMT1 messenger RNA, DNMT1 protein, and global DNA methylation levels in the SCNT embryos. Meanwhile, the survival rate after electrofusion (90.4% vs. 89.4% vs. 89.1%, P > 0.05) and developmental rates of the SCNT embryos (72.8% vs. 74.9% vs. 72.4%, P > 0.05; 29.7% vs. 31.7% vs. 29.7%, P > 0.05) were not significantly affected. In summary, siRNAs were effectively transfected into the SCNT embryos via the proposed method and exert their functions, and the normal development of preimplantation SCNT embryos was not affected by the method used.
Collapse
Affiliation(s)
- Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - LiJun Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - WenZhe Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - QingFu Mao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - YongSheng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qian Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Song Hua
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
39
|
Egea RR, Puchalt NG, Escrivá MM, Varghese AC. OMICS: Current and future perspectives in reproductive medicine and technology. J Hum Reprod Sci 2014; 7:73-92. [PMID: 25191020 PMCID: PMC4150148 DOI: 10.4103/0974-1208.138857] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/14/2014] [Accepted: 02/26/2014] [Indexed: 12/16/2022] Open
Abstract
Many couples present fertility problems at their reproductive age, and although in the last years, the efficiency of assisted reproduction techniques has increased, these are still far from being 100% effective. A key issue in this field is the proper assessment of germ cells, embryos and endometrium quality, in order to determine the actual likelihood to succeed. Currently available analysis is mainly based on morphological features of oocytes, sperm and embryos and although these strategies have improved the results, there is an urgent need of new diagnostic and therapeutic tools. The emergence of the - OMICS technologies (epigenomics, genomics, transcriptomics, proteomics and metabolomics) permitted the improvement on the knowledge in this field, by providing with a huge amount of information regarding the biological processes involved in reproductive success, thereby getting a broader view of complex biological systems with a relatively low cost and effort.
Collapse
Affiliation(s)
- Rocío Rivera Egea
- Andrology Laboratory and Semen Bank, Instituto Universitario, IVI Valencia, Spain
| | | | | | | |
Collapse
|
40
|
Phua DCY, Xu J, Ali SM, Boey A, Gounko NV, Hunziker W. ZO-1 and ZO-2 are required for extra-embryonic endoderm integrity, primitive ectoderm survival and normal cavitation in embryoid bodies derived from mouse embryonic stem cells. PLoS One 2014; 9:e99532. [PMID: 24905925 PMCID: PMC4048262 DOI: 10.1371/journal.pone.0099532] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/15/2014] [Indexed: 12/14/2022] Open
Abstract
The Zonula Occludens proteins ZO-1 and ZO-2 are cell-cell junction-associated adaptor proteins that are essential for the structural and regulatory functions of tight junctions in epithelial cells and their absence leads to early embryonic lethality in mouse models. Here, we use the embryoid body, an in vitro peri-implantation mouse embryogenesis model, to elucidate and dissect the roles ZO-1 and ZO-2 play in epithelial morphogenesis and de novo tight junction assembly. Through the generation of individual or combined ZO-1 and ZO-2 null embryoid bodies, we show that their dual deletion prevents tight junction formation, resulting in the disorganization and compromised barrier function of embryoid body epithelial layers. The disorganization is associated with poor microvilli development, fragmented basement membrane deposition and impaired cavity formation, all of which are key epithelial tissue morphogenetic processes. Expression of Podocalyxin, which positively regulates the formation of microvilli and the apical membrane, is repressed in embryoid bodies lacking both ZO-1 and ZO-2 and this correlates with an aberrant submembranous localization of Ezrin. The null embryoid bodies thus give an insight into how the two ZO proteins influence early mouse embryogenesis and possible mechanisms underlying the embryonic lethal phenotype.
Collapse
Affiliation(s)
- Dominic C. Y. Phua
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jianliang Xu
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Safiah Mohamed Ali
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Adrian Boey
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- IMB-IMCB Joint Electron Microscopy Suite, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Natalia V. Gounko
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- IMB-IMCB Joint Electron Microscopy Suite, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Walter Hunziker
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Department of Physiology, National University of Singapore and Singapore Eye Research Institute (SERI), Singapore, Singapore
- * E-mail:
| |
Collapse
|
41
|
Yin S, Wu H, Lv J, Wu X, Zhang Y, Du J, Zhang Y. SHP-1 arrests mouse early embryo development through downregulation of Nanog by dephosphorylation of STAT3. PLoS One 2014; 9:e86330. [PMID: 24466030 PMCID: PMC3897670 DOI: 10.1371/journal.pone.0086330] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/07/2013] [Indexed: 01/14/2023] Open
Abstract
Src-homology protein tyrosine phosphatase-1 (SHP-1) is a protein tyrosine phosphatase that is implicated in the regulation of growth, differentiation, survival, apoptosis and proliferation of hematopoietic cells and other cell types. Here, we found that SHP-1 is involved in regulation of early embryonic development. Embryos overexpressing SHP-1 were mainly arrested at the 8-cell stage, and Nanog mRNA expression was first observed in the morulae that showed down-regulation of SHP-1. These results suggested an antagonistic relationship between SHP-1 and Nanog during early embryonic development. Next, the specific mechanism was examined in mouse F9 embryonal carcinoma cells. We confirmed that signal transducer and activator of transcription 3 (STAT3) was a substrate for SHP-1 by co-immunoprecipitation. Using overexpression and knockdown strategies, we found that SHP-1 participated in regulation of Nanog expression. Furthermore, site mutation of STAT3 was performed to confirm that SHP-1 was responsible for rapid STAT3 dephosphorylation and a decrease of Nanog expression in F9 cells. These findings suggest that SHP-1 plays a crucial role during early embryonic development. Thus, SHP-1 may function as a key regulator for Nanog that specifically demarcates the nascent epiblast, coincident with the domain of X chromosome reprogramming.
Collapse
Affiliation(s)
- Songna Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Haibo Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiaxing Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinying Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Yan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Juan Du
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail:
| |
Collapse
|
42
|
Protein O-mannosylation is crucial for E-cadherin-mediated cell adhesion. Proc Natl Acad Sci U S A 2013; 110:21024-9. [PMID: 24297939 DOI: 10.1073/pnas.1316753110] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In recent years protein O-mannosylation has become a focus of attention as a pathomechanism underlying severe congenital muscular dystrophies associated with neuronal migration defects. A key feature of these disorders is the lack of O-mannosyl glycans on α-dystroglycan, resulting in abnormal basement membrane formation. Additional functions of O-mannosylation are still largely unknown. Here, we identify the essential cell-cell adhesion glycoprotein epithelial (E)-cadherin as an O-mannosylated protein and establish a functional link between O-mannosyl glycans and cadherin-mediated cell-cell adhesion. By genetically and pharmacologically blocking protein O-mannosyltransferases, we found that this posttranslational modification is essential for preimplantation development of the mouse embryo. O-mannosylation-deficient embryos failed to proceed from the morula to the blastocyst stage because of defects in the molecular architecture of cell-cell contact sites, including the adherens and tight junctions. Using mass spectrometry, we demonstrate that O-mannosyl glycans are present on E-cadherin, the major cell-adhesion molecule of blastomeres, and present evidence that this modification is generally conserved in cadherins. Further, the use of newly raised antibodies specific for an O-mannosyl-conjugated epitope revealed that these glycans are present on early mouse embryos. Finally, our cell-aggregation assays demonstrated that O-mannosyl glycans are crucial for cadherin-based cell adhesion. Our results redefine the significance of O-mannosylation in humans and other mammals, showing the immense impact of cadherins on normal as well as pathogenic cell behavior.
Collapse
|
43
|
|
44
|
Developmental expression and possible functional roles of mouse Nlrp4e in preimplantation embryos. In Vitro Cell Dev Biol Anim 2013; 49:548-53. [PMID: 23708922 DOI: 10.1007/s11626-013-9638-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 05/12/2013] [Indexed: 12/27/2022]
Abstract
Increasing evidence suggests that some Nlrp genes are crucial for oogenesis, folliculogenesis, and early embryonic development. Nlrp4e is one of seven copies of Nlrp4, which plays a putative role in the reproduction system in mice. Gene duplication is regarded as an important driving force behind the evolution of novel genes with new or altered functions. We investigated the role of Nlrp4e in oocyte and preimplantation embryos by determining its expression profile using quantitative real-time polymerase chain reaction. Nlrp4e mRNA accumulated during oogenesis. Moreover, Nlrp4e transcripts were upregulated during the two-cell stage and then declined sharply and became almost undetectable, which represents a crucial time for major embryonic genome activation in the mouse. Knockdown of Nlrp4e in fertilized eggs using RNA interference resulted in arrested development between the two- and eight-cell stages in a dose-dependent manner. However, targeted inhibition of Nlrp4e in germinal-vesicle-stage oocytes had no phenotypic effects on oocyte maturation. The above experiments were also carried out in parthenogenetic embryos to determine the effects of Nlrp4e in embryos without a paternal genome. The results of this study indicate that Nlrp4e, a maternal-zygotic-effect gene, may not be involved in oocyte maturation but may play a critical role in early embryogenesis.
Collapse
|
45
|
Peng H, Wu Y, Zhang Y. Efficient delivery of DNA and morpholinos into mouse preimplantation embryos by electroporation. PLoS One 2012; 7:e43748. [PMID: 22928027 PMCID: PMC3424252 DOI: 10.1371/journal.pone.0043748] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/23/2012] [Indexed: 01/19/2023] Open
Abstract
Mouse preimplantation development is characterized by three major transitions and two lineage segregations. Each transition or lineage segregation entails pronounced changes in the pattern of gene expression. Thus, research into the function of genes with obvious changes in expression pattern will shed light on the molecular basis of preimplantation development. We have described a simplified and effective method–electroporation–of introducing plasmid DNA and morpholinos into mouse preimplantation embryos and verified effectiveness of this approach by testing the procedure on the endogenous gene Oct4. Before electroporation, the zona pellucida was weakened by the treatment of acid Tyrode’s solution. Then we optimized the parameters such as voltage, pulse duration, number of pulses and repeats, and applied these parameters to subsequent experiments. Compared with the control groups, the number of apoptotic cells and the expression and localization of OCT3/4 or CDX2 was not significantly changed in blastocysts developed from 1-cell embryos, which were electroporated with pIRES2-AcGFP1-Nuc eukaryotic expression vector or mismatched morpholino oligonucleotides. Furthermore, electroporated plasmid DNA and morpholinos targeting the endogenous gene Oct4 were able to sharply down regulate expression of OCT4 protein and actually cause expected phenotypes in mouse preimplantation embryos. In conclusion, plasmid DNA and morpholinos could be efficient delivered into mouse preimplantation embryos by electroporation and exert their functions, and normal development of preimplantation embryos was not affected.
Collapse
Affiliation(s)
- Hui Peng
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People’s Republic of China
| | - Yongyan Wu
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People’s Republic of China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People’s Republic of China
- * E-mail:
| |
Collapse
|
46
|
Syed ZA, Bougé AL, Byri S, Chavoshi TM, Tång E, Bouhin H, van Dijk-Härd IF, Uv A. A luminal glycoprotein drives dose-dependent diameter expansion of the Drosophila melanogaster hindgut tube. PLoS Genet 2012; 8:e1002850. [PMID: 22876194 PMCID: PMC3410870 DOI: 10.1371/journal.pgen.1002850] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 06/06/2012] [Indexed: 11/19/2022] Open
Abstract
An important step in epithelial organ development is size maturation of the organ lumen to attain correct dimensions. Here we show that the regulated expression of Tenectin (Tnc) is critical to shape the Drosophila melanogaster hindgut tube. Tnc is a secreted protein that fills the embryonic hindgut lumen during tube diameter expansion. Inside the lumen, Tnc contributes to detectable O-Glycans and forms a dense striated matrix. Loss of tnc causes a narrow hindgut tube, while Tnc over-expression drives tube dilation in a dose-dependent manner. Cellular analyses show that luminal accumulation of Tnc causes an increase in inner and outer tube diameter, and cell flattening within the tube wall, similar to the effects of a hydrostatic pressure in other systems. When Tnc expression is induced only in cells at one side of the tube wall, Tnc fills the lumen and equally affects all cells at the lumen perimeter, arguing that Tnc acts non-cell-autonomously. Moreover, when Tnc expression is directed to a segment of a tube, its luminal accumulation is restricted to this segment and affects the surrounding cells to promote a corresponding local diameter expansion. These findings suggest that deposition of Tnc into the lumen might contribute to expansion of the lumen volume, and thereby to stretching of the tube wall. Consistent with such an idea, ectopic expression of Tnc in different developing epithelial tubes is sufficient to cause dilation, while epidermal Tnc expression has no effect on morphology. Together, the results show that epithelial tube diameter can be modelled by regulating the levels and pattern of expression of a single luminal glycoprotein. Epithelial tubes constitute the functional units of vital organs, and they undergo highly regulated changes in size and shape during development to accommodate the three-dimensional configurations optimal for organ physiology. Through studies of Drosophila melanogaster, we show that epithelial tube diameter can be modelled simply by regulating the levels and pattern of expression of a single glycoprotein. The protein is secreted into the tubular lumen, where it forms a dense matrix and acts in a dose-dependent manner to drive diameter growth. We suggest that deposition of the protein into the lumen promotes local expansion of the lumen volume, and thereby stretching of the surrounding tube wall. Such a mechanism could represent a general means to adjust tube diameter during epithelial organ development.
Collapse
Affiliation(s)
- Zulfeqhar A. Syed
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anne-Laure Bougé
- Centre des Sciences du Goût et de l'Alimentation, Université de Bourgogne, Dijon, France
| | - Sunitha Byri
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Tina M. Chavoshi
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Erika Tång
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Hervé Bouhin
- Centre des Sciences du Goût et de l'Alimentation, Université de Bourgogne, Dijon, France
| | - Iris F. van Dijk-Härd
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- * E-mail: (AU), (IFvD-H)
| | - Anne Uv
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- * E-mail: (AU), (IFvD-H)
| |
Collapse
|
47
|
Liu WM, Pang RTK, Cheong AWY, Ng EHY, Lao K, Lee KF, Yeung WSB. Involvement of microRNA lethal-7a in the regulation of embryo implantation in mice. PLoS One 2012; 7:e37039. [PMID: 22623977 PMCID: PMC3356405 DOI: 10.1371/journal.pone.0037039] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 04/12/2012] [Indexed: 01/23/2023] Open
Abstract
MicroRNAs interact with multiple mRNAs resulting in their degradation and/or translational repression. This report used the delayed implantation model to determine the role of miRNAs in blastocysts. Dormant blastocysts in delayed implanting mice were activated by estradiol. Differential expression of 45 out of 238 miRNAs examined was found between the dormant and the activated blastocysts. Five of the nine members of the microRNA lethal-7 (let-7) family were down-regulated after activation. Human blastocysts also had a low expression of let-7 family. Forced-expression of a family member, let-7a in mouse blastocysts decreased the number of implantation sites (let-7a: 1.1±0.4; control: 3.8±0.4) in vivo, and reduced the percentages of blastocyst that attached (let-7a: 42.0±8.3%; control: 79.0±5.1%) and spreaded (let-7a: 33.5±2.9%; control: 67.3±3.8%) on fibronectin in vitro. Integrin-β3, a known implantation-related molecule, was demonstrated to be a target of let-7a by 3′-untranslated region reporter assay in cervical cancer cells HeLa, and Western blotting in mouse blastocysts. The inhibitory effect of forced-expression of let-7a on blastocyst attachment and outgrowth was partially nullified in vitro and in vivo by forced-expression of integrin-β3. This study provides the first direct evidence that let-7a is involved in regulating the implantation process partly via modulation of the expression of integrin-β3. (200 words).
Collapse
Affiliation(s)
- Wei-Min Liu
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
| | - Ronald T. K. Pang
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
- Centre for Reproduction, Development and Growth, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
| | - Ana W. Y. Cheong
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
| | - Kaiqin Lao
- Applied Biosystems, Foster City, California, United States of America
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
- Centre for Reproduction, Development and Growth, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
- Centre for Reproduction, Development and Growth, The University of Hong Kong, Pokfulam, Hong Kong, People’s Republic of China
- * E-mail:
| |
Collapse
|
48
|
Mendoza-Naranjo A, Cormie P, Serrano AE, Hu R, O'Neill S, Wang CM, Thrasivoulou C, Power KT, White A, Serena T, Phillips ARJ, Becker DL. Targeting Cx43 and N-cadherin, which are abnormally upregulated in venous leg ulcers, influences migration, adhesion and activation of Rho GTPases. PLoS One 2012; 7:e37374. [PMID: 22615994 PMCID: PMC3352877 DOI: 10.1371/journal.pone.0037374] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 04/18/2012] [Indexed: 12/19/2022] Open
Abstract
Background Venous leg ulcers can be very hard to heal and represent a significant medical need with no effective therapeutic treatment currently available. Principal Findings In wound edge biopsies from human venous leg ulcers we found a striking upregulation of dermal N-cadherin, Zonula Occludens-1 and the gap junction protein Connexin43 (Cx43) compared to intact skin, and in stark contrast to the down-regulation of Cx43 expression seen in acute, healing wounds. We targeted the expression of these proteins in 3T3 fibroblasts to evaluate their role in venous leg ulcers healing. Knockdown of Cx43 and N-cadherin, but not Zonula Occludens-1, accelerated cell migration in a scratch wound-healing assay. Reducing Cx43 increased Golgi reorientation, whilst decreasing cell adhesion and proliferation. Furthermore, Connexin43 and N-cadherin knockdown led to profound effects on fibroblast cytoskeletal dynamics after scratch-wounding. The cells exhibited longer lamelipodial protrusions lacking the F-actin belt seen at the leading edge in wounded control cells. This phenotype was accompanied by augmented activation of Rac-1 and RhoA GTPases, as revealed by Förster Resonance Energy Transfer and pull down experiments. Conclusions Cx43 and N-cadherin are potential therapeutic targets in the promotion of healing of venous leg ulcers, by acting at least in part through distinct contributions of cell adhesion, migration, proliferation and cytoskeletal dynamics.
Collapse
Affiliation(s)
- Ariadna Mendoza-Naranjo
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
- * E-mail: (DLB); (AMN)
| | - Peter Cormie
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Antonio E. Serrano
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rebecca Hu
- CoDa Therapeutics, Auckland, New Zealand
| | | | - Chiuhui Mary Wang
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | | - Kieran T. Power
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | | - Thomas Serena
- Newbridge Medical Research Corp, Warren, Pennsylvania, United States of America
| | | | - David L. Becker
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
- * E-mail: (DLB); (AMN)
| |
Collapse
|
49
|
Acloque H, Ocaña OH, Nieto MA. Mutual exclusion of transcription factors and cell behaviour in the definition of vertebrate embryonic territories. Curr Opin Genet Dev 2012; 22:308-14. [PMID: 22560468 DOI: 10.1016/j.gde.2012.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/07/2012] [Accepted: 03/18/2012] [Indexed: 12/27/2022]
Abstract
Early embryonic territories are transient entities under permanent remodelling to form newly derived cell populations that will eventually give rise to the adult tissues and organs. A vast effort has been devoted to identifying the determinants and mechanisms that define embryonic territories. Indeed, studies in the vertebrate embryo from the morula stage to the segregation of the main embryonic layers-ectoderm, mesoderm and endoderm-have highlighted the importance of the mutual exclusion/repression between pairs of transcription factors, in coordination with the control exerted over cell division, adhesion and motility.
Collapse
Affiliation(s)
- Hervé Acloque
- Instituto de Neurociencias CSIC-UMH, Avda Ramón y Cajal s/n, San Juan de Alicante, 03550, Spain; UMR 444, INRA-ENVT, Génétique Cellulaire, Toulouse, France
| | | | | |
Collapse
|
50
|
Hur CG, Kim EJ, Cho SK, Cho YW, Yoon SY, Tak HM, Kim CW, Choe C, Han J, Kang D. K+ efflux through two-pore domain K+ channels is required for mouse embryonic development. Reproduction 2012; 143:625-36. [DOI: 10.1530/rep-11-0225] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Numerous studies have suggested that K+ channels regulate a wide range of physiological processes in mammalian cells. However, little is known about the specific function of K+ channels in germ cells. In this study, mouse zygotes were cultured in a medium containing K+ channel blockers to identify the functional role of K+ channels in mouse embryonic development. Voltage-dependent K+ channel blockers, such as tetraethylammonium and BaCl2, had no effect on embryonic development to the blastocyst stage, whereas K2P channel blockers, such as quinine, selective serotonin reuptake inhibitors (fluoxetine, paroxetine, and citalopram), gadolinium trichloride, anandamide, ruthenium red, and zinc chloride, significantly decreased blastocyst formation (P<0.05). RT-PCR data showed that members of the K2P channel family, specifically KCNK2, KCNK10, KCNK4, KCNK3, and KCNK9, were expressed in mouse oocytes and embryos. In addition, their mRNA expression levels, except Kcnk3, were up-regulated by above ninefold in morula-stage embryos compared with 2-cell stage embryos (2-cells). Immunocytochemical data showed that KCNK2, KCNK10, KCNK4, KCNK3, and KCNK9 channel proteins were expressed in the membrane of oocytes, 2-cells, and blastocysts. Each siRNA injection targeted at Kcnk2, Kcnk10, Kcnk4, Kcnk3, and Kcnk9 significantly decreased blastocyst formation by ∼38% compared with scrambled siRNA injection (P<0.05). The blockade of K2P channels acidified the intracellular pH and depolarized the membrane potential. These results suggest that K2P channels could improve mouse embryonic development through the modulation of gating by activators.
Collapse
|