1
|
Petersen M, Reyes-Vigil F, Campo M, Brusés JL. Classical cadherins evolutionary constraints in primates is associated with their expression in the central nervous system. PLoS One 2024; 19:e0313428. [PMID: 39570883 PMCID: PMC11581309 DOI: 10.1371/journal.pone.0313428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Classical cadherins (CDH) comprise a family of single-pass transmembrane glycoproteins that contribute to tissue morphogenesis by regulating cell-cell adhesion, cytoskeletal dynamics, and cell signaling. CDH are grouped into type I (CDH 1, 2, 3, 4 and 15) and type II (CDH 5, 6, 7, 8, 9, 10, 11, 12, 18, 20, 22 and 24), based on the folding of the cadherin binding domain involved in trans-dimer formation. CDH are exclusively found in metazoans, and the origin and expansion of the gene family coincide with the emergence of multicellularity and vertebrates respectively. This study examined the evolutionary changes of CDH orthologs in primates and the factors that influence selective pressure to investigate the varying constraints exerted among CDH. Pairwise comparisons of the number of amino acid substitutions and of the ratio of non-synonymous substitutions per non-synonymous sites (dN) over synonymous substitutions per synonymous sites (dS), show that CDH2, CDH4, and most type II CDH have been under significantly higher negative selective pressure as compared to CDH1, CDH3, CDH5 and CDH19. Evaluation of gene essentiality as determined by the effect of germline deletion on animal viability, morphogenic phenotype, and reproductive fitness, show no correlation with the with extent of negative selection observed on CDH. Spearman's correlation analysis shows a positive correlation between CDH expression levels (E) in mouse and human tissues and their rate of evolution (R), as observed in most proteins expressed on the cell surface. However, CDH expression in the CNS show a significant E-R negative correlation, indicating that the strong negative selection exerted on CDH2, CDH4, and most type II CDH is associated with their expression in the CNS. CDH participate in a variety of cellular processes in the CNS including neuronal migration and functional assembly of neural circuits, which could profoundly influence animal fitness. Therefore, our findings suggest that the unusually high negative selective pressure exerted on CDH2, CDH4 and most type II CDH is due to their role in CNS formation and function and may have contributed to shape the evolution of the CNS in primates.
Collapse
Affiliation(s)
- Max Petersen
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Fredy Reyes-Vigil
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Marc Campo
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Juan L. Brusés
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| |
Collapse
|
2
|
Schliffka MF, Dumortier JG, Pelzer D, Mukherjee A, Maître JL. Inverse blebs operate as hydraulic pumps during mouse blastocyst formation. Nat Cell Biol 2024; 26:1669-1677. [PMID: 39261717 DOI: 10.1038/s41556-024-01501-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
During preimplantation development, mouse embryos form a fluid-filled lumen. Pressurized fluid fractures cell-cell contacts and accumulates into pockets, which coarsen into a single lumen. How the embryo controls intercellular fluid movement during coarsening is unknown. Here we report inverse blebs growing into cells at adhesive contacts. Throughout the embryo we observed hundreds of inverse blebs, each filling with intercellular fluid and retracting within a minute. Inverse blebs grow due to pressure build-up resulting from fluid accumulation and cell-cell adhesion, which locally confines fluid. Inverse blebs retract due to actomyosin contraction, practically pushing fluid within the intercellular space. Importantly, inverse blebs occur infrequently at contacts formed by multiple cells, which effectively serve as fluid sinks. Manipulation of the embryo topology reveals that without sinks inverse blebs pump fluid into one another in futile cycles. We propose that inverse blebs operate as hydraulic pumps to promote luminal coarsening, thereby constituting an instrument used by cells to control fluid movement.
Collapse
Affiliation(s)
- Markus F Schliffka
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
- Carl Zeiss SAS, Marly-le-Roy, France
| | - Julien G Dumortier
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
| | - Diane Pelzer
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
| | - Arghyadip Mukherjee
- Laboratoire de physique de l'École Normale Supérieure, CNRS UMR 8023, PSL Research University, Sorbonne Université and Université Paris Cité, Paris, France.
| | - Jean-Léon Maître
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France.
| |
Collapse
|
3
|
Zhang Z, Zhanghuang C, Mi T, Jin L, Liu J, Li M, Wu X, Wang J, Li M, Wang Z, Guo P, He D. The PI3K-AKT-mTOR signaling pathway mediates the cytoskeletal remodeling and epithelial-mesenchymal transition in bladder outlet obstruction. Heliyon 2023; 9:e21281. [PMID: 38027933 PMCID: PMC10663759 DOI: 10.1016/j.heliyon.2023.e21281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Partial bladder outlet obstruction(pBOO) is the most common cause of lower urinary tract symptoms (LUTS) and significantly affects the quality of life. Long-term pBOO can cause changes in bladder structure and function, referred to as bladder remodeling. The pathogenesis of pBOO-induced bladder remodeling has yet to be fully understood, so effective treatment options are lacking. Our study aimed to explore how pBOO-induced bladder remodeling brings new strategies for treating pBOO. Methods A rat model of pBOO was established by partial ligation of the bladder neck, and the morphological changes and fibrosis changes in the bladder tissues were detected by H&E and Masson trichrome staining. Furthermore, EMT(epithelial-mesenchymal transition) related indicators and related pathway changes were further examined after TGF- β treatment of urothelial cells SV-HUC-1. Finally, the above indicators were tested again after using the PI3K inhibitor. Subsequently, RNA sequencing of bladder tissues to identify differential genes and related pathways enrichment and validated by immunofluorescence and western blotting analysis. Results The pBOO animal model was successfully established by partially ligating the bladder neck. H&E staining showed significant changes in the bladder structure, and Masson trichrome staining showed significantly increased collagen fibers. RNA sequencing results significantly enriched in the cytoskeleton, epithelial-mesenchymal transformation, and the PI3K-AKT-mTOR signaling pathway. Immunofluorescence and western blotting revealed EMT and cytoskeletal remodeling in SV-HUC-1 cells after induction of TGF- β and in the pBOO bladder tissues. The western blotting showed significant activation of the PI3K-AKT-mTOR signaling pathway in SV-HUC-1 cells after induction of TGF-β and in pBOO bladder tissues. Furthermore, EMT and cytoskeletal damage were partially reversed after PI3K pathway inhibition using PI3K inhibitors. Conclusions In the pBOO rat model, the activation of the PI3K-AKT-mTOR signaling pathway can mediate the cytoskeletal remodeling and the EMT to induce fibrosis in the bladder tissues. PI3K inhibitors partially reversed EMT and cytoskeletal damage.
Collapse
Affiliation(s)
- Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Chenghao Zhanghuang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
- Department of Urology, Children's Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650103, PR China
| | - Tao Mi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Liming Jin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Jiayan Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Maoxian Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Xin Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Jinkui Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Mujie Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Zhang Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Peng Guo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China
- China International Science and Technology Cooperation Base of Child Development and Critical, National Clinical Research Center for Child Health and Disorders, Chongqing, PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| |
Collapse
|
4
|
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
|
5
|
Sathyanarayanan A, Ing-Simmons E, Chen R, Jeong HW, Ozguldez HO, Fan R, Duethorn B, Kim KP, Kim YS, Stehling M, Brinkmann H, Schöler HR, Adams RH, Vaquerizas JM, Bedzhov I. Early developmental plasticity enables the induction of an intermediate extraembryonic cell state. SCIENCE ADVANCES 2022; 8:eabl9583. [PMID: 36332016 PMCID: PMC9635831 DOI: 10.1126/sciadv.abl9583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/19/2022] [Indexed: 05/23/2023]
Abstract
Two fundamental elements of pre-implantation embryogenesis are cells' intrinsic self-organization program and their developmental plasticity, which allows embryos to compensate for alterations in cell position and number; yet, these elements are still poorly understood. To be able to decipher these features, we established culture conditions that enable the two fates of blastocysts' extraembryonic lineages-the primitive endoderm and the trophectoderm-to coexist. This plasticity emerges following the mechanisms of the first lineage segregation in the mouse embryo, and it manifests as an extended potential for extraembryonic chimerism during the pre-implantation embryogenesis. Moreover, this shared state enables robust assembly into higher-order blastocyst-like structures, thus combining both the cell fate plasticity and self-organization features of the early extraembryonic lineages.
Collapse
Affiliation(s)
- Anusha Sathyanarayanan
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Elizabeth Ing-Simmons
- Regulatory Genomics Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Rui Chen
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster, Germany
- Faculty of Medicine, University of Münster, Röntgenstrasse 20, 48149 Münster, Germany
| | - Hatice O. Ozguldez
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Rui Fan
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Binyamin Duethorn
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero Seocho-gu, Seoul 06591, Korea
| | - Yung Su Kim
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Heike Brinkmann
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hans R. Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Ralf H. Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster, Germany
- Faculty of Medicine, University of Münster, Röntgenstrasse 20, 48149 Münster, Germany
| | - Juan M. Vaquerizas
- Regulatory Genomics Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Ivan Bedzhov
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| |
Collapse
|
6
|
Gerber TS, Ridder DA, Schindeldecker M, Weinmann A, Duret D, Breuhahn K, Galle PR, Schirmacher P, Roth W, Lang H, Straub BK. Constitutive Occurrence of E:N-cadherin Heterodimers in Adherens Junctions of Hepatocytes and Derived Tumors. Cells 2022; 11:cells11162507. [PMID: 36010583 PMCID: PMC9406782 DOI: 10.3390/cells11162507] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022] Open
Abstract
Cell–cell junctions are pivotal for embryogenesis and tissue homeostasis but also play a major role in tumorigenesis, tumor invasion, and metastasis. E-cadherin (CDH1) and N-cadherin (CDH2) are two adherens junction’s transmembrane glycoproteins with tissue-specific expression patterns in epithelial and neural/mesenchymal cells. Aberrant expression has been implicated in the process of epithelial–mesenchymal transition (EMT) in malignant tumors. We could hitherto demonstrate cis-E:N-cadherin heterodimer in endoderm-derived cells. Using immunoprecipitation in cultured cells of the line PLC as well as in human hepatocellular carcinoma (HCC)-lysates, we isolated E-N-cadherin heterodimers in a complex with the plaque proteins α- and β-catenin, plakoglobin, and vinculin. In confocal laser scanning microscopy, E-cadherin co-localized with N-cadherin at the basolateral membrane of normal hepatocytes, hepatocellular adenoma (HCA), and in most cases of HCC. In addition, we analyzed E- and N-cadherin expression via immunohistochemistry in a large cohort of 868 HCCs from 570 patients, 25 HCA, and respective non-neoplastic liver tissue, and correlated our results with multiple prognostic markers. While E- or N-cadherin were similarly expressed in tumor sites with vascular invasion or HCC metastases, HCC with vascular encapsulated tumor clusters (VETC) displayed slightly reduced E-cadherin, and slightly increased N-cadherin expression. Analyzing The Cancer Genome Atlas patient cohort, we found that reduced mRNA levels of CDH1, but not CDH2 were significantly associated with unfavorable prognosis; however, in multivariate analysis, CDH1 did not correlate with prognosis. In summary, E- and N-cadherin are specific markers for hepatocytes and derived HCA and HCC. E:N-cadherin heterodimers are constitutively expressed in the hepatocytic lineage and only slightly altered in malignant progression, thereby not complying with the concept of EMT.
Collapse
Affiliation(s)
- Tiemo Sven Gerber
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Dirk Andreas Ridder
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Mario Schindeldecker
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Tissue Biobank, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Arndt Weinmann
- 1st Department of Internal Medicine, Gastroenterology and Hepatology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Diane Duret
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Peter R. Galle
- 1st Department of Internal Medicine, Gastroenterology and Hepatology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Beate Katharina Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Correspondence:
| |
Collapse
|
7
|
Hawdon A, Aberkane A, Zenker J. Microtubule-dependent subcellular organisation of pluripotent cells. Development 2021; 148:272646. [PMID: 34710215 DOI: 10.1242/dev.199909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
With the advancement of cutting-edge live imaging technologies, microtubule remodelling has evolved as an integral regulator for the establishment of distinct differentiated cells. However, despite their fundamental role in cell structure and function, microtubules have received less attention when unravelling the regulatory circuitry of pluripotency. Here, we summarise the role of microtubule organisation and microtubule-dependent events required for the formation of pluripotent cells in vivo by deciphering the process of early embryogenesis: from fertilisation to blastocyst. Furthermore, we highlight current advances in elucidating the significance of specific microtubule arrays in in vitro culture systems of pluripotent stem cells and how the microtubule cytoskeleton serves as a highway for the precise intracellular movement of organelles. This Review provides an informed understanding of the intrinsic role of subcellular architecture of pluripotent cells and accentuates their regenerative potential in combination with innovative light-inducible microtubule techniques.
Collapse
Affiliation(s)
- Azelle Hawdon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Asma Aberkane
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jennifer Zenker
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
8
|
Punovuori K, Malaguti M, Lowell S. Cadherins in early neural development. Cell Mol Life Sci 2021; 78:4435-4450. [PMID: 33796894 PMCID: PMC8164589 DOI: 10.1007/s00018-021-03815-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 11/12/2022]
Abstract
During early neural development, changes in signalling inform the expression of transcription factors that in turn instruct changes in cell identity. At the same time, switches in adhesion molecule expression result in cellular rearrangements that define the morphology of the emerging neural tube. It is becoming increasingly clear that these two processes influence each other; adhesion molecules do not simply operate downstream of or in parallel with changes in cell identity but rather actively feed into cell fate decisions. Why are differentiation and adhesion so tightly linked? It is now over 60 years since Conrad Waddington noted the remarkable "Constancy of the Wild Type" (Waddington in Nature 183: 1654-1655, 1959) yet we still do not fully understand the mechanisms that make development so reproducible. Conversely, we do not understand why directed differentiation of cells in a dish is sometimes unpredictable and difficult to control. It has long been suggested that cells make decisions as 'local cooperatives' rather than as individuals (Gurdon in Nature 336: 772-774, 1988; Lander in Cell 144: 955-969, 2011). Given that the cadherin family of adhesion molecules can simultaneously influence morphogenesis and signalling, it is tempting to speculate that they may help coordinate cell fate decisions between neighbouring cells in the embryo to ensure fidelity of patterning, and that the uncoupling of these processes in a culture dish might underlie some of the problems with controlling cell fate decisions ex-vivo. Here we review the expression and function of cadherins during early neural development and discuss how and why they might modulate signalling and differentiation as neural tissues are formed.
Collapse
Affiliation(s)
- Karolina Punovuori
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, 00290, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Mattias Malaguti
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sally Lowell
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
9
|
Ikuta R, Myoenzono K, Wasano J, Hamaguchi-Hamada K, Hamada S, Kurumata-Shigeto M. N-cadherin localization in taste buds of mouse circumvallate papillae. J Comp Neurol 2020; 529:2227-2242. [PMID: 33319419 DOI: 10.1002/cne.25090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/03/2023]
Abstract
Taste buds, the receptor organs for taste, contain 50-100 taste bud cells. Although these cells undergo continuous turnover, the structural and functional integrity of taste buds is maintained. The molecular mechanisms by which synaptic connectivity between taste buds and afferent fibers is formed and maintained remain ambiguous. In the present study, we examined the localization of N-cadherin in the taste buds of the mouse circumvallate papillae because N-cadherin, one of the classical cadherins, is important for the formation and maintenance of synapses. At the light microscopic level, N-cadherin was predominantly detected in type II cells and nerve fibers in the connective tissues in and around the vallate papillae. At the ultrastructural level, N-cadherin immunoreactivity appears along the cell membrane and in the intracellular vesicles of type II cells. N-cadherin immunoreactivity also is evident in the membranes of afferent terminals at the contact sites to N-cadherin-positive type II cells. At channel type synapses between type II cells and nerve fibers, N-cadherin is present surrounding, but not within, the presumed neurotransmitter release zone, identified by large mitochondria apposed to the taste cells. The present results suggest that N-cadherin is important for the formation or maintenance of type II cell afferent synapses in taste buds.
Collapse
Affiliation(s)
- Rio Ikuta
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Kanae Myoenzono
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan.,Humanome Lab., Inc., Tokyo, Japan
| | - Jun Wasano
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | | | - Shun Hamada
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Mami Kurumata-Shigeto
- International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| |
Collapse
|
10
|
Zhou W, Santos L, Dimitriadis E. Characterization of the role for cadherin 6 in the regulation of human endometrial receptivity. Reprod Biol Endocrinol 2020; 18:66. [PMID: 32600462 PMCID: PMC7322878 DOI: 10.1186/s12958-020-00624-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The endometrial luminal epithelium is the first point of attachment of embryos during implantation. Failure of embryos to firmly adhere results in implantation failure and infertility. A receptive endometrial luminal epithelium is achieved through the expression of adhesion molecules in the mid-secretory phase and is a requirement for implantation. Cadherin 6 (CDH6) is an adhesion molecule localizing to the endometrial luminal epithelial cell surface in the mid-secretory/receptive phase and knockdown of CDH6 in the Ishikawa cells (receptive endometrial epithelial cell line) compromises cell integrity. However, there are no studies investigating the role of CDH6 on receptivity and infertility. This study aimed to investigate whether CDH6 is dysregulated in the endometrium of women with infertility during the receptive window and the effect of CDH6 on endometrial adhesion and receptivity. METHODS The expression and the localization of CDH6 in the human endometrium were determined by immunohistochemistry. Ishikawa cells were used to investigate the functional consequences of CDH6 knockdown on endometrial adhesive capacity to HTR8/SVneo (trophoblast cell line) spheroids in vitro. CDH6 knockdown was assessed by qPCR and immunoblotting. After CDH6 knockdown, the expression of type II cadherin family members and CDH6 functional partners were assessed by qPCR. Two-tailed unpaired student's t-test or one-way ANOVA as appropriate were used for statistical analysis with a significance threshold of P < 0.05. RESULTS A significant reduction of CDH6 immunolocalization was recorded in the luminal and glandular epithelium of endometrium from women with infertility (P < 0.05) compared to fertile group respective cellular compartments in the mid-secretory phase. Functional analysis using Ishikawa cells demonstrated that knockdown of CDH6 (treated with 50 nM CDH6 siRNA) significantly reduced epithelial adhesive capacity (P < 0.05) to HTR8/SVneo spheroids compared to control and other type II cadherin family members likely failed to compensate for the loss of CDH6. The expression levels of CDH6 functional partners, catenin family members were not changed after CDH6 knockdown in Ishikawa cells. CONCLUSION Together, our data revealed that CDH6 was dysregulated in the endometrium from women with infertility and altered Ishikawa cell adhesive capacity. Our study supports a role for CDH6 in regulating endometrial adhesion and implantation.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| | - Leilani Santos
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia.
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
11
|
D'Occhio MJ, Campanile G, Zicarelli L, Visintin JA, Baruselli PS. Adhesion molecules in gamete transport, fertilization, early embryonic development, and implantation-role in establishing a pregnancy in cattle: A review. Mol Reprod Dev 2020; 87:206-222. [PMID: 31944459 DOI: 10.1002/mrd.23312] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
Cell-cell adhesion molecules have critically important roles in the early events of reproduction including gamete transport, sperm-oocyte interaction, embryonic development, and implantation. Major adhesion molecules involved in reproduction include cadherins, integrins, and disintegrin and metalloprotease domain-containing (ADAM) proteins. ADAMs on the surface of sperm adhere to integrins on the oocyte in the initial stages of sperm-oocyte interaction and fusion. Cadherins act in early embryos to organize the inner cell mass and trophectoderm. The trophoblast and uterine endometrial epithelium variously express cadherins, integrins, trophinin, and selectin, which achieve apposition and attachment between the elongating conceptus and uterine epithelium before implantation. An overview of the major cell-cell adhesion molecules is presented and this is followed by examples of how adhesion molecules help shape early reproductive events. The argument is made that a deeper understanding of adhesion molecules and reproduction will inform new strategies that improve embryo survival and increase the efficiency of natural mating and assisted breeding in cattle.
Collapse
Affiliation(s)
- Michael J D'Occhio
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Luigi Zicarelli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - José A Visintin
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Pietro S Baruselli
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
12
|
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
|
13
|
Punovuori K, Migueles RP, Malaguti M, Blin G, Macleod KG, Carragher NO, Pieters T, van Roy F, Stemmler MP, Lowell S. N-cadherin stabilises neural identity by dampening anti-neural signals. Development 2019; 146:dev.183269. [PMID: 31601548 PMCID: PMC6857587 DOI: 10.1242/dev.183269] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022]
Abstract
A switch from E- to N-cadherin regulates the transition from pluripotency to neural identity, but the mechanism by which cadherins regulate differentiation was previously unknown. Here, we show that the acquisition of N-cadherin stabilises neural identity by dampening anti-neural signals. We use quantitative image analysis to show that N-cadherin promotes neural differentiation independently of its effects on cell cohesiveness. We reveal that cadherin switching diminishes the level of nuclear β-catenin, and that N-cadherin also dampens FGF activity and consequently stabilises neural fate. Finally, we compare the timing of cadherin switching and differentiation in vivo and in vitro, and find that this process becomes dysregulated during in vitro differentiation. We propose that N-cadherin helps to propagate a stable neural identity throughout the emerging neuroepithelium, and that dysregulation of this process contributes to asynchronous differentiation in culture. Summary: As pluripotent cells undergo neural differentiation they swap E-cadherin for N-cadherin. This switch in adhesion molecules modulates signalling in order to facilitate the differentiation process.
Collapse
Affiliation(s)
- Karolina Punovuori
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rosa P Migueles
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Guillaume Blin
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Kenneth G Macleod
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Neil O Carragher
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Tim Pieters
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Center for Medical Genetics, Ghent University Hospital; Cancer Research Institute Ghent (CRIG), Ghent B-9000, Belgium
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Cancer Research Institute Ghent (CRIG), Ghent B-9000, Belgium
| | - Marc P Stemmler
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
14
|
Pei D, Shu X, Gassama-Diagne A, Thiery JP. Mesenchymal–epithelial transition in development and reprogramming. Nat Cell Biol 2019; 21:44-53. [DOI: 10.1038/s41556-018-0195-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023]
|
15
|
Barui A, Datta P. Biophysical factors in the regulation of asymmetric division of stem cells. Biol Rev Camb Philos Soc 2018; 94:810-827. [PMID: 30467934 DOI: 10.1111/brv.12479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Ananya Barui
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| | - Pallab Datta
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| |
Collapse
|
16
|
Liu S, Bou G, Zhao J, Guo S, Guo J, Weng X, Yin Z, Liu Z. Asynchronous CDX2 expression and polarization of porcine trophoblast cells reflects a species-specific trophoderm lineage determination progress model. Mol Reprod Dev 2018; 85:590-598. [PMID: 29719081 DOI: 10.1002/mrd.22994] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/01/2018] [Indexed: 11/10/2022]
Abstract
Upregulation of Cdx2 expression in outer cells is a key event responsible for cell lineage segregation between the inner cell mass and the trophoderm (TE) in mouse morula-stage embryos. In TE cells, polarization can regulate Hippo and Rho-associated kinase (Rho-ROCK) signaling to induce the nuclear location of YAP, which has been demonstrated to further induce the expression of Cdx2. However, we found that CDX2 expression could not be detected in the outer cells of porcine morula-stage embryos but only in some TE cells at the early blastocyst stage. The biological significance and the regulation mechanism of this species-specific CDX2 expression pattern have still not been determined. We show here that an asynchronous CDX2 expression pattern exists in porcine TE cells during the development of the blastocyst. We demonstrate that CDX2 expression in porcine TE cells depends on the nuclear localization of YAP and polarization of the embryo through Y27632 treatment. We found that the polarization process in the morula to the late blastocyst stage porcine embryos was asynchronous, which was revealed by the apical localization of phosphorylated EZRIN staining. Artificially enhancing the number of polarized blastomeres by culturing the separated blastomeres of four-cell stage porcine embryos resulted in increased CDX2-positive cell numbers. These results indicate that the mechanism of CDX2 expression regulation is conserved, but the polarization progress is not conserved between the pig and the mouse, and results in a species-specific trophoblast determination progress model.
Collapse
Affiliation(s)
- Shichao Liu
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Gerelchimeg Bou
- Animal Science College, Inner Mongolia Agricultural University, Hohhot, China
| | - Jianchao Zhao
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Shimeng Guo
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Jia Guo
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Xiaogang Weng
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Zhi Yin
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| |
Collapse
|
17
|
Tatapudy S, Aloisio F, Barber D, Nystul T. Cell fate decisions: emerging roles for metabolic signals and cell morphology. EMBO Rep 2017; 18:2105-2118. [PMID: 29158350 DOI: 10.15252/embr.201744816] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/14/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022] Open
Abstract
Understanding how cell fate decisions are regulated is a fundamental goal of developmental and stem cell biology. Most studies on the control of cell fate decisions address the contributions of changes in transcriptional programming, epigenetic modifications, and biochemical differentiation cues. However, recent studies have found that other aspects of cell biology also make important contributions to regulating cell fate decisions. These cues can have a permissive or instructive role and are integrated into the larger network of signaling, functioning both upstream and downstream of developmental signaling pathways. Here, we summarize recent insights into how cell fate decisions are influenced by four aspects of cell biology: metabolism, reactive oxygen species (ROS), intracellular pH (pHi), and cell morphology. For each topic, we discuss how these cell biological cues interact with each other and with protein-based mechanisms for changing gene transcription. In addition, we highlight several questions that remain unanswered in these exciting and relatively new areas of the field.
Collapse
Affiliation(s)
- Sumitra Tatapudy
- Departments of Anatomy and OB-GYN/RS, University of California, San Francisco, San Francisco, CA, USA
| | - Francesca Aloisio
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Diane Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Todd Nystul
- Departments of Anatomy and OB-GYN/RS, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Segal JM, Ward CM. Novel peptides for deciphering structural and signalling functions of E-cadherin in mouse embryonic stem cells. Sci Rep 2017; 7:41827. [PMID: 28169326 PMCID: PMC5294416 DOI: 10.1038/srep41827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/28/2016] [Indexed: 01/21/2023] Open
Abstract
We have previously shown that E-cadherin regulates the naive pluripotent state of mouse embryonic stem cells (mESCs) by enabling LIF-dependent STAT3 phosphorylation, with E-cadherin null mESCs exhibiting over 3000 gene transcript alterations and a switch to Activin/Nodal-dependent pluripotency. However, elucidation of the exact mechanisms associated with E-cadherin function in mESCs is compounded by the difficulty in delineating the structural and signalling functions of this protein. Here we show that mESCs treated with the E-cadherin neutralising antibody DECMA-1 or the E-cadherin binding peptide H-SWELYYPLRANL-NH2 (Epep) exhibit discrete profiles for pluripotent transcripts and NANOG protein expression, demonstrating that the type of E-cadherin inhibitor employed dictates the cellular phenotype of mESCs. Alanine scanning mutation of Epep revealed residues critical for Tbx3, Klf4 and Esrrb transcript repression, cell-cell contact abrogation, cell survival in suspension, STAT3 phosphorylation and water solubility. STAT3 phosphorylation was found to be independent of loss of cell-cell contact and Activin/Nodal-dependent pluripotency and a peptide is described that enhances STAT3 phosphorylation and Nanog transcript and protein expression in mESCs. These peptides represent a useful resource for deciphering the structural and signalling functions of E-cadherin and demonstrate that complete absence of E-cadherin protein is likely required for hierarchical signalling pathway alterations in mESCs.
Collapse
Affiliation(s)
- Joe M. Segal
- Stem Cell Research Group, The University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Christopher M. Ward
- Stem Cell Research Group, The University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
19
|
Sauvegarde C, Paul D, Bridoux L, Jouneau A, Degrelle S, Hue I, Rezsohazy R, Donnay I. Dynamic Pattern of HOXB9 Protein Localization during Oocyte Maturation and Early Embryonic Development in Mammals. PLoS One 2016; 11:e0165898. [PMID: 27798681 PMCID: PMC5087947 DOI: 10.1371/journal.pone.0165898] [Citation(s) in RCA: 6] [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: 07/01/2016] [Accepted: 10/01/2016] [Indexed: 02/06/2023] Open
Abstract
Background We previously showed that the homeodomain transcription factor HOXB9 is expressed in mammalian oocytes and early embryos. However, a systematic and exhaustive study of the localization of the HOXB9 protein, and HOX proteins in general, during mammalian early embryonic development has so far never been performed. Results The distribution of HOXB9 proteins in oocytes and the early embryo was characterized by immunofluorescence from the immature oocyte stage to the peri-gastrulation period in both the mouse and the bovine. HOXB9 was detected at all studied stages with a dynamic expression pattern. Its distribution was well conserved between the two species until the blastocyst stage and was mainly nuclear. From that stage on, trophoblastic cells always showed a strong nuclear staining, while the inner cell mass and the derived cell lines showed important dynamic variations both in staining intensity and in intra-cellular localization. Indeed, HOXB9 appeared to be progressively downregulated in epiblast cells and only reappeared after gastrulation had well progressed. The protein was also detected in the primitive endoderm and its derivatives with a distinctive presence in apical vacuoles of mouse visceral endoderm cells. Conclusions Together, these results could suggest the existence of unsuspected functions for HOXB9 during early embryonic development in mammals.
Collapse
Affiliation(s)
- Caroline Sauvegarde
- Biologie Moléculaire et Cellulaire Animale (AMCB), Institut des Sciences de la Vie (ISV), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Delphine Paul
- Biologie Moléculaire et Cellulaire Animale (AMCB), Institut des Sciences de la Vie (ISV), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Laure Bridoux
- Biologie Moléculaire et Cellulaire Animale (AMCB), Institut des Sciences de la Vie (ISV), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Alice Jouneau
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - Séverine Degrelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S1139, U767, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- PremUp Foundation, Paris, France
| | - Isabelle Hue
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - René Rezsohazy
- Biologie Moléculaire et Cellulaire Animale (AMCB), Institut des Sciences de la Vie (ISV), Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Isabelle Donnay
- Biologie Moléculaire et Cellulaire Animale (AMCB), Institut des Sciences de la Vie (ISV), Université catholique de Louvain, Louvain-la-Neuve, Belgium
- * E-mail:
| |
Collapse
|
20
|
Leung CY, Zhu M, Zernicka-Goetz M. Polarity in Cell-Fate Acquisition in the Early Mouse Embryo. Curr Top Dev Biol 2016; 120:203-34. [PMID: 27475853 DOI: 10.1016/bs.ctdb.2016.04.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Establishing polarity is a fundamental part of embryogenesis and can be traced back to the earliest developmental stages. It can be achieved in one of two ways: through the preexisting polarization of germ cells before fertilization or via symmetry breaking after fertilization. In mammals, it seems to be the latter, and we will discuss the various cytological and molecular events that lead up to this event, its mechanisms and the consequences. In mammals, the first polarization event occurs in the preimplantation period, when the embryo is but a cluster of cells, free-floating in the oviduct. This provides a unique, autonomous system to study the de novo polarization that is essential to life. In this review, we will cover modern and past studies on the polarization of the early embryo, using the mouse as a model system, as well as hypothesizing the potential implications and functions of the biological events involved.
Collapse
Affiliation(s)
- C Y Leung
- University of Cambridge, Cambridge, United Kingdom
| | - M Zhu
- University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
21
|
Basilicata MF, Frank M, Solter D, Brabletz T, Stemmler MP. Inappropriate cadherin switching in the mouse epiblast compromises proper signaling between the epiblast and the extraembryonic ectoderm during gastrulation. Sci Rep 2016; 6:26562. [PMID: 27217206 PMCID: PMC4877576 DOI: 10.1038/srep26562] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/05/2016] [Indexed: 11/09/2022] Open
Abstract
Cadherin switching from E-cadherin (E-cad) to N-cadherin (N-cad) is a key step of the epithelial-mesenchymal transition (EMT) processes that occurs during gastrulation and cancer progression. We investigate whether cadherins actively participate in progression of EMT by crosstalk to signaling pathways. We apply ectopic cadherin switching before the onset of mouse gastrulation. Mutants with an induced E-cad to N-cad switch (Ncadki) die around E8.5. Severe morphological changes including a small epiblast, a rounded shape, an enlarged extra-embryonic compartment and lack of the amnion, combined with a massive cell detachment from the ectodermal layer are detected. In contrast to epiblast-specific E-cad depletion, gastrulation is initiated in Ncadki embryos, but patterning of the germ-layers is abnormal. An overall reduction in BMP signaling, expansion of Nodal and Eomes domains, combined with reduced Wnt3a expression at the primitive streak is observed. Our results show that in addition to cadherin-dependent adhesion, proper embryonic development requires E-cad mediated signaling function to facilitate a feedback loop that stabilizes Bmp4 and Bmp2 expression in the extraembryonic ectoderm and sustained downstream activity in the epiblast. Moreover, for proper morphogenesis a fine-tuned spatio-temporal control of cadherin switching is required during EMT at gastrulation to avoid premature cell detachment and migration.
Collapse
Affiliation(s)
- M Felicia Basilicata
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Marcus Frank
- Electron Microscopy Center, University Medicine Rostock, Strempelstr. 14, 18057 Rostock, Germany
| | - Davor Solter
- Epithelial Epigenetics and Development Lab, Institute of Medical Biology, A*STAR, Singapore
| | - Thomas Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, University of Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| | - Marc P Stemmler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany.,Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, University of Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| |
Collapse
|
22
|
Inada M, Izawa G, Kobayashi W, Ozawa M. 293 cells express both epithelial as well as mesenchymal cell adhesion molecules. Int J Mol Med 2016; 37:1521-7. [PMID: 27121032 PMCID: PMC4866952 DOI: 10.3892/ijmm.2016.2568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/24/2016] [Indexed: 11/06/2022] Open
Abstract
The 293 cell line, used extensively in various types of studies due to the ease with which these cells can be transfected, was thought to be derived by the transformation of primary cultures of human embryonic kidney cells with sheared adenovirus type 5 DNA. Although the 293 cells were assumed to originate from epithelial cells, the exact origin of these cells remains unknown. Previous attempts to characterize these cells combined immunostaining, immunoblot analysis and microarray analysis to demonstrate that 293 cells express neurofilament subunits, α-internexin, and several other proteins typically found in neurons. These findings raised the possibility that the 293 cell line may have originated from human neuronal lineage cells. Contrary to this suggestion, in this study, we found that the 293 cells expressed N-cadherin and vimentin, which are marker proteins expressed in mesenchymal cells. Furthermore, the 293 cells also expressed E-cadherin, cytokeratins 5/8 and desmoglein 2, which are epithelial cell markers. When the cells, primarily cultured from the kidneys of Clawn miniature swine and passaged 10–15 generations [termed porcine kidney epithelial (PKE) cells] were examined, they were found to be positive for the expression of both mesenchymal and epithelial markers. Thus, transformation by adenovirus was not necessary for the cells to express N-cadherin. Occludin and zonula occludens (ZO)-1, two components of tight junctions in epithelial and endothelial cells, were detected in the 293 and the PKE cells. Thus, the findings of the present study demonstrate that 293 cells retain several characteristics of epithelial cells.
Collapse
Affiliation(s)
- Masakazu Inada
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Genya Izawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Wakako Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
23
|
Lee YL, Fong SW, Chen AC, Li T, Yue C, Lee CL, Ng EH, Yeung WS, Lee KF. Establishment of a novel human embryonic stem cell-derived trophoblastic spheroid implantation model. Hum Reprod 2015; 30:2614-26. [DOI: 10.1093/humrep/dev223] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022] Open
|
24
|
Hawkins K, Keramari M, Soncin F, Segal JM, Mohamet L, Miazga N, Ritson S, Bobola N, Merry CLR, Ward CM. Novel cell lines isolated from mouse embryonic stem cells exhibiting de novo methylation of the E-cadherin promoter. Stem Cells 2015; 32:2869-79. [PMID: 25074424 DOI: 10.1002/stem.1790] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/04/2014] [Accepted: 06/07/2014] [Indexed: 12/25/2022]
Abstract
Mouse embryonic stem cells (mESCs) and epiblast stem cells represent the naïve and primed pluripotent states, respectively. These cells self-renew via distinct signaling pathways and can transition between the two states in the presence of appropriate growth factors. Manipulation of signaling pathways has therefore allowed the isolation of novel pluripotent cell types such as Fibroblast growth factor, Activin and BIO-derived stem cells and IESCs. However, the effect of cell seeding density on pluripotency remains unexplored. In this study, we have examined whether mESCs can epigenetically regulate E-cadherin to enter a primed-like state in response to low cell seeding density. We show that low density seeding in the absence of leukaemia inhibitory factor (LIF) induces decreased apoptosis and maintenance of pluripotency via Activin/Nodal, concomitant with loss of E-cadherin, Signal transducer and activator of transcription phosphorylation, and chimera-forming ability. These cells, E-cadherin negative proliferating stem cells (ENPSCs) can be reverted to a naïve phenotype by addition of LIF or forced E-cadherin expression. However, prolonged culture of ENPSCs without LIF leads to methylation of the E-cadherin promoter (ENPSC(M)), which cannot be reversed by LIF supplementation, and increased histone H3K27 and decreased H3K4 trimethylation. Transcript analysis of ENPSC(M) revealed a primed-like phenotype and their differentiation leads to enrichment of neuroectoderm cells. The generation of ENPSCs is similar to tumorigenesis as ENPSCs exhibit transcript alterations associated with neoplasia, hyperplasia, carcinoma, and metastasis. We therefore describe a novel cell model to elucidate the role of E-cadherin in pluripotency and to investigate epigenetic regulation of this gene during mESC differentiation and tumor metastasis.
Collapse
Affiliation(s)
- Kate Hawkins
- Stem Cell Research Group, The University of Manchester, Core Technology Facility, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Farouz Y, Chen Y, Terzic A, Menasché P. Concise Review: Growing Hearts in the Right Place: On the Design of Biomimetic Materials for Cardiac Stem Cell Differentiation. Stem Cells 2015; 33:1021-35. [DOI: 10.1002/stem.1929] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Yohan Farouz
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
| | - Yong Chen
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
| | | | - Philippe Menasché
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Department of Cardiovascular Surgery; Paris France
| |
Collapse
|
26
|
Applying the proximity ligation assay (PLA) to mouse preimplantation embryos for identifying protein-protein interactions in situ. Methods Mol Biol 2015; 1233:57-64. [PMID: 25319889 DOI: 10.1007/978-1-4939-1789-1_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Analysis of protein-protein interactions in mouse preimplantation embryos is hindered by the low cell number of the embryo. Here we describe the use of the proximity ligation assay to overcome these limitations and outline how protein-protein interactions can be visualized in situ. The method is based on a normal immunofluorescence labeling protocol of preimplantation embryos. Events of binding of the two primary antibodies directed against two individual proteins close to each other are visualized. If the two primary antibodies and the corresponding oligo-linked secondary antibodies bind in close proximity a cascade of events is initiated. This includes oligo ligation, DNA amplification, and hybridization with a fluorescent probe that allows visualization of this close proximity. In contrast to normal immunofluorescence labeling, here detection of red fluorescent dots reflects protein-protein interaction.
Collapse
|
27
|
Schneider MR, Kolligs FT. E-cadherin's role in development, tissue homeostasis and disease: Insights from mouse models: Tissue-specific inactivation of the adhesion protein E-cadherin in mice reveals its functions in health and disease. Bioessays 2014; 37:294-304. [PMID: 25449798 DOI: 10.1002/bies.201400141] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent studies uncovered critical roles of the adhesion protein E-cadherin in health and disease. Global inactivation of Cdh1, the gene encoding E-cadherin in mice, results in early embryonic lethality due to an inability to form the trophectodermal epithelium. To unravel E-cadherin's functions beyond development, numerous mouse lines with tissue-specific disruption of Cdh1 have been generated. The consequences of E-cadherin loss showed great variability depending on the tissue in question, ranging from nearly undetectable changes to a complete loss of tissue structure and function. This review focuses on these studies and discusses how they provided important insights into E-cadherin's role in cell adhesion, proliferation and differentiation, and its consequences for biological processes as epithelial-to-mesenchymal transition, vascularization, and carcinogenesis. Lastly, we present some perspectives and possible approaches for future research.
Collapse
Affiliation(s)
- Marlon R Schneider
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Germany
| | | |
Collapse
|
28
|
Lang RA, Herman K, Reynolds AB, Hildebrand JD, Plageman TF. p120-catenin-dependent junctional recruitment of Shroom3 is required for apical constriction during lens pit morphogenesis. Development 2014; 141:3177-87. [PMID: 25038041 DOI: 10.1242/dev.107433] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Apical constriction (AC) is a widely utilized mechanism of cell shape change whereby epithelial cells transform from a cylindrical to conical shape, which can facilitate morphogenetic movements during embryonic development. Invertebrate epithelial cells undergoing AC depend on the contraction of apical cortex-spanning actomyosin filaments that generate force on the apical junctions and pull them toward the middle of the cell, effectively reducing the apical circumference. A current challenge is to determine whether these mechanisms are conserved in vertebrates and to identify the molecules responsible for linking apical junctions with the AC machinery. Utilizing the developing mouse eye as a model, we have uncovered evidence that lens placode AC may be partially dependent on apically positioned myosin-containing filaments associated with the zonula adherens. In addition we found that, among several junctional components, p120-catenin genetically interacts with Shroom3, a protein required for AC during embryonic morphogenesis. Further analysis revealed that, similar to Shroom3, p120-catenin is required for AC of lens cells. Finally, we determined that p120-catenin functions by recruiting Shroom3 to adherens junctions. Together, these data identify a novel role for p120-catenin during AC and further define the mechanisms required for vertebrate AC.
Collapse
Affiliation(s)
- Richard A Lang
- The Visual System Group, Division of Pediatric Ophthalmology and Developmental Biology, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ken Herman
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey D Hildebrand
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Timothy F Plageman
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
29
|
Pieters T, van Roy F. Role of cell–cell adhesion complexes in embryonic stem cell biology. J Cell Sci 2014; 127:2603-13. [DOI: 10.1242/jcs.146720] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Pluripotent embryonic stem cells (ESCs) can self-renew or differentiate into any cell type within an organism. Here, we focus on the roles of cadherins and catenins – their cytoplasmic scaffold proteins – in the fate, maintenance and differentiation of mammalian ESCs. E-cadherin is a master stem cell regulator that is required for both mouse ESC (mESC) maintenance and differentiation. E-cadherin interacts with key components of the naive stemness pathway and ablating it prevents stem cells from forming well-differentiated teratomas or contributing to chimeric animals. In addition, depleting E-cadherin converts naive mouse ESCs into primed epiblast-like stem cells (EpiSCs). In line with this, a mesenchymal-to-epithelial transition (MET) occurs during reprogramming of somatic cells towards induced pluripotent stem cells (iPSCs), leading to downregulation of N-cadherin and acquisition of high E-cadherin levels. β-catenin exerts a dual function; it acts in cadherin-based adhesion and in WNT signaling and, although WNT signaling is important for stemness, the adhesive function of β-catenin might be crucial for maintaining the naive state of stem cells. In addition, evidence is rising that other junctional proteins are also important in ESC biology. Thus, precisely regulated levels and activities of several junctional proteins, in particular E-cadherin, safeguard naive pluripotency and are a prerequisite for complete somatic cell reprogramming.
Collapse
Affiliation(s)
- Tim Pieters
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
- Molecular and Cellular Oncology Unit, Inflammation Research Center, VIB, B-9052 Ghent, Belgium
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
- Molecular Cell Biology Unit, Inflammation Research Center, VIB, B-9052 Ghent, Belgium
| |
Collapse
|
30
|
Engl W, Arasi B, Yap LL, Thiery JP, Viasnoff V. Actin dynamics modulate mechanosensitive immobilization of E-cadherin at adherens junctions. Nat Cell Biol 2014; 16:587-94. [DOI: 10.1038/ncb2973] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 04/24/2014] [Indexed: 02/06/2023]
|
31
|
The States of Pluripotency: Pluripotent Lineage Development in the Embryo and in the Dish. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/208067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The pluripotent cell lineage of the embryo comprises a series of temporally and functionally distinct intermediary cell states, the epiblast precursor cell of the newly formed blastocyst, the epiblast population of the inner cell mass, and the early and late epiblast of the postimplantation embryo, referred to here as early and late primitive ectoderm. Pluripotent cell populations representative of the embryonic populations can be formed in culture. Although multiple pluripotent cell states are now recognised, little is known about the signals and pathways that progress cells from the epiblast precursor cell to the late primitive ectoderm in the embryo or in culture. The characterisation of cell states is most advanced in mouse where conditions for culturing distinct pluripotent cell states are well established and embryonic material is accessible. This review will focus on the pluripotent cell states present during embryonic development in the mouse and what is known of the mechanisms that regulate the progression of the lineage from the epiblast precursor cell and the ground state of pluripotency to the late primitive ectoderm present immediately prior to cell differentiation.
Collapse
|
32
|
Hu A, Shang C, Li Q, Sun N, Wu L, Ma Y, Jiao X, Min J, Zeng G, He X. Epithelial-mesenchymal transition delayed by E-cad to promote tissue formation in hepatic differentiation of mouse embryonic stem cells in vitro. Stem Cells Dev 2014; 23:877-87. [PMID: 24266635 DOI: 10.1089/scd.2013.0354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hepatic differentiation of embryonic stem cells (ESCs) usually results in a single cell lineage, and the formation of liver tissues remains difficult. Here, we examine the role of epithelial-mesenchymal transition (EMT) that is regulated by epithelial cadherin (E-cad) expression in hepatic tissue formation from ESCs. E-cad was transfected into mouse ESCs to enable a stable expression of E-cad. Hepatic differentiation of ESCs was then induced by hepatic growth factors. Wnt/β-catenin signaling and EMT speed were examined to determine the differentiation process. Hepatic and angiogenesis markers, as well as differentiated cell-adhesive force were also examined to identify the hepatic tissue differentiation. In our results, E-cad expression gradually decreased in normal ESC (N-ESC) differentiation, but remained stable in the E-cad transfected ESC (EC-ESC) group. In EC-ESC differentiation, expressions of cytoplastic β-catenin and EMT were much lower and significantly prolonged. Angiogenesis markers vascular endothelial growth factor receptor-1 (VEGFR-1) and CD31/PECAM-1 were expressed only on day 5-13 in N-ESC differentiation, whereas VEGFR-1 and CD31/PECAM-1 were expressed prolonged on day 5-17 in the EC-ESC group and were coincident with the expression of hepatic markers. Finally, EC-ESC differentiation maintained multilayer-growth patterns, and abundant vascular network structures appeared and migrated in albumin-positive cell areas. The cellular adhesion forces between embryonic body cells in EC-ESC differentiation during day 13-17 were similar to those of mouse liver tissue. In conclusion, accelerated EMT due to the decreased E-cad expression may partially contribute to the failure of hepatic tissue formation in N-ESC differentiation. E-cad can act in synergy with hepatic growth factors and facilitate the early-stage formation of hepatic tissues through down-regulating Wnt/β-catenin signaling and delaying EMT. This work provides a new insight into hepatic tissue differentiation that is mediated by E-cad from ESC.
Collapse
Affiliation(s)
- Anbin Hu
- 1 Department of General Surgery, The First Affiliated Hospital, Sun Yat-sen University , Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
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
|
35
|
Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:317-36. [PMID: 23481201 DOI: 10.1016/b978-0-12-394311-8.00014-5] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a process whereby epithelial cells are transcriptionally reprogrammed, resulting in decreased adhesion and enhanced migration or invasion. EMT occurs during different stages of embryonic development, including gastrulation and neural crest cell delamination, and is induced by a panel of specific transcription factors. These factors comprise, among others, members of the Snail, ZEB, and Twist families, and are all known to modulate cadherin expression and, in particular, E-cadherin. By regulating expression of the cadherin family of proteins, EMT-inducing transcription factors dynamically modulate cell adhesion, allowing many developmental processes to take place. However, during cancer progression EMT can be utilized by cancer cells to contribute to malignancy. This is also reflected at the level of the cadherins, where the cadherin switch between E- and N-cadherins is a classical example seen in cancer-related EMT. In this chapter, we give a detailed overview of the entanglement between EMT-inducing transcription factors and cadherin modulation during embryonic development and cancer progression. We describe how classical cadherins such as E- and N-cadherins are regulated during EMT, as well as cadherin 7, -6B, and -11.
Collapse
Affiliation(s)
- Alexander Gheldof
- Department for Molecular Biomedical Research, Unit of Molecular and Cellular Oncology, VIB, Ghent, Belgium
| | | |
Collapse
|
36
|
Adhesion, but not a specific cadherin code, is indispensable for ES cell and induced pluripotency. Stem Cell Res 2013; 11:1250-63. [DOI: 10.1016/j.scr.2013.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 06/24/2013] [Accepted: 08/17/2013] [Indexed: 10/26/2022] Open
|
37
|
Itoh Y, Tyssowski K, Gotoh Y. Transcriptional coupling of neuronal fate commitment and the onset of migration. Curr Opin Neurobiol 2013; 23:957-64. [PMID: 23973158 DOI: 10.1016/j.conb.2013.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 12/18/2022]
Abstract
During mammalian CNS development, when the neural precursor cells commit to the neuronal fate they must delaminate and migrate toward the pial surface in order to reach the appropriate final location. Thus, the coordination of delamination and fate commitment is important in creating the correct structure. Although previous studies have proposed that spindle orientation during mitosis plays a role in both delamination and fate commitment, thus coordinating these events, subsequent studies have challenged this model. Recent work has identified several transcriptional mechanisms associated with neurogenesis that inhibit cell adhesion of newborn neurons and intermediate neuronal progenitors, thereby triggering delamination and linking it with fate commitment.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | | | | |
Collapse
|
38
|
McPherson NO, Bakos HW, Owens JA, Setchell BP, Lane M. Improving metabolic health in obese male mice via diet and exercise restores embryo development and fetal growth. PLoS One 2013; 8:e71459. [PMID: 23977045 PMCID: PMC3747240 DOI: 10.1371/journal.pone.0071459] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 07/03/2013] [Indexed: 12/17/2022] Open
Abstract
Paternal obesity is now clearly associated with or causal of impaired embryo and fetal development and reduced pregnancy rates in humans and rodents. This appears to be a result of reduced blastocyst potential. Whether these adverse embryo and fetal outcomes can be ameliorated by interventions to reduce paternal obesity has not been established. Here, male mice fed a high fat diet (HFD) to induce obesity were used, to determine if early embryo and fetal development is improved by interventions of diet (CD) and/or exercise to reduce adiposity and improve metabolism. Exercise and to a lesser extent CD in obese males improved embryo development rates, with increased cell to cell contacts in the compacting embryo measured by E-cadherin in exercise interventions and subsequently, increased blastocyst trophectoderm (TE), inner cell mass (ICM) and epiblast cell numbers. Implantation rates and fetal development from resulting blastocysts were also improved by exercise in obese males. Additionally, all interventions to obese males increased fetal weight, with CD alone and exercise alone, also increasing fetal crown-rump length. Measures of embryo and fetal development correlated with paternal measures of glycaemia, insulin action and serum lipids regardless of paternal adiposity or intervention, suggesting a link between paternal metabolic health and subsequent embryo and fetal development. This is the first study to show that improvements to metabolic health of obese males through diet and exercise can improve embryo and fetal development, suggesting such interventions are likely to improve offspring health.
Collapse
Affiliation(s)
- Nicole O. McPherson
- School of Paediatrics and Reproductive Health, Discipline of Obstetrics and Gynaecology, University of Adelaide, South Australia, Australia
- * E-mail:
| | - Hassan W. Bakos
- School of Medicine, Discipline of Medicine, University of Adelaide, South Australia, Australia
- Repromed, Dulwich, South Australia, Australia
| | - Julie A. Owens
- School of Paediatrics and Reproductive Health, Discipline of Obstetrics and Gynaecology, University of Adelaide, South Australia, Australia
| | - Brian P. Setchell
- School of Medical Sciences, Discipline of Anatomy, University of Adelaide, South Australia, Australia
| | - Michelle Lane
- School of Paediatrics and Reproductive Health, Discipline of Obstetrics and Gynaecology, University of Adelaide, South Australia, Australia
- Repromed, Dulwich, South Australia, Australia
| |
Collapse
|
39
|
Nekrasova O, Green KJ. Desmosome assembly and dynamics. Trends Cell Biol 2013; 23:537-46. [PMID: 23891292 DOI: 10.1016/j.tcb.2013.06.004] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 01/06/2023]
Abstract
Desmosomes are intercellular junctions that anchor intermediate filaments (IFs) to the plasma membrane, forming a supracellular scaffold that provides mechanical resilience to tissues. This anchoring function is accomplished by specialized members of the cadherin family and associated cytoskeletal linking proteins, which together form a highly organized membrane core flanked by mirror-image cytoplasmic plaques. Due to the biochemical insolubility of desmosomes, the mechanisms that govern assembly of these components into a functional organelle remained elusive. Recently developed molecular reporters and live cell imaging approaches have provided powerful new tools to monitor this finely tuned process in real time. Here we discuss studies that are beginning to decipher the machinery and regulation governing desmosome assembly and homeostasis in situ and how these mechanisms are affected during disease pathogenesis.
Collapse
Affiliation(s)
- Oxana Nekrasova
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
40
|
Radice GL. N-cadherin-mediated adhesion and signaling from development to disease: lessons from mice. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:263-89. [PMID: 23481199 PMCID: PMC6047516 DOI: 10.1016/b978-0-12-394311-8.00012-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Of the 20 classical cadherin subtypes identified in mammals, the functions of the two initially identified family members E- (epithelial) and N- (neural) cadherin have been most extensively studied. E- and N-Cadherin have mostly mutually exclusive expression patterns, with E-cadherin expressed primarily in epithelial cells, whereas N-cadherin is found in a variety of cells, including neural, muscle, and mesenchymal cells. N-Cadherin function, in particular, appears to be cell context-dependent, as it can mediate strong cell-cell adhesion in the heart but induces changes in cell behavior in favor of a migratory phenotype in the context of epithelial-mesenchymal transition (EMT). The ability of tumor cells to alter their cadherin expression profile, for example, E- to N-cadherin, is critical for malignant progression. Recent advances in mouse molecular genetics, and specifically tissue-specific knockout and knockin alleles of N-cadherin, have provided some unexpected results. This chapter highlights some of the genetic studies that explored the complex role of N-cadherin in embryonic development and disease.
Collapse
Affiliation(s)
- Glenn L Radice
- Department of Medicine, Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Hashimoto T, Soeno Y, Maeda G, Taya Y, Aoba T, Nasu M, Kawashiri S, Imai K. Progression of oral squamous cell carcinoma accompanied with reduced E-cadherin expression but not cadherin switch. PLoS One 2012; 7:e47899. [PMID: 23110125 PMCID: PMC3479144 DOI: 10.1371/journal.pone.0047899] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 09/18/2012] [Indexed: 01/28/2023] Open
Abstract
The cadherin switch from E-cadherin to N-cadherin is considered as a hallmark of the epithelial-mesenchymal transition and progression of carcinomas. Although it enhances aggressive behaviors of adenocarcinoma cells, the significance and role of cadherin switch in squamous cell carcinomas (SCCs) are largely controversial. In the present study, we immunohistochemically examined expression of E-cadherin and N-cadherin in oral SCCs (n = 63) and its implications for the disease progression. The E-cadherin-positive carcinoma cells were rapidly decreased at the invasive front. The percentage of carcinoma cells stained E-cadherin at the cell membrane was reduced in parallel with tumor dedifferentiation (P<0.01) and enhanced invasion (P<0.01). In contrast, N-cadherin-positive cells were very limited and did not correlate with the clinicopathological parameters. Mouse tongue tumors xenotransplantated oral SCC cell lines expressing both cadherins in vitro reproduced the reduction of E-cadherin-positive carcinoma cells at the invasive front and the negligible expression of N-cadherin. These results demonstrate that the reduction of E-cadherin-mediated carcinoma cell-cell adhesion at the invasive front, but not the cadherin switch, is an important determinant for oral SCC progression, and suggest that the environments surrounding carcinoma cells largely affect the cadherin expression.
Collapse
Affiliation(s)
- Takashi Hashimoto
- Department of Biochemistry, The Nippon Dental University, Tokyo, Japan
| | - Yuichi Soeno
- Department of Pathology, The Nippon Dental University, Tokyo, Japan
| | - Genta Maeda
- Department of Biochemistry, The Nippon Dental University, Tokyo, Japan
| | - Yuji Taya
- Department of Pathology, The Nippon Dental University, Tokyo, Japan
| | - Takaaki Aoba
- Department of Pathology, The Nippon Dental University, Tokyo, Japan
| | - Masanori Nasu
- Research Center for Odontology, The Nippon Dental University, Tokyo, Japan
| | | | - Kazushi Imai
- Department of Biochemistry, The Nippon Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
42
|
Kohan-Ghadr HR, Smith LC, Arnold DR, Murphy BD, Lefebvre RC. Aberrant expression of E-cadherin and β-catenin proteins in placenta of bovine embryos derived from somatic cell nuclear transfer. Reprod Fertil Dev 2012; 24:588-98. [PMID: 22541547 DOI: 10.1071/rd11162] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/27/2011] [Indexed: 12/11/2022] Open
Abstract
Abnormal placental development is common in the bovine somatic cell nuclear transfer (SCNT)-derived fetus. In the present study, we characterised the expression of E-cadherin and β-catenin, structural proteins of adherens junctions, in SCNT gestations as a model for impaired placentation. Cotyledonary tissues were separated from pregnant uteri of SCNT (n = 6) and control pregnancies (n = 8) obtained by artificial insemination. Samples were analysed by western blot, quantitative RT-PCR (qRT-PCR) and immunohistochemistry. Bovine trophectoderm cell lines derived from SCNT and control embryos were analysed to compare with the in utero condition. Although no differences in E-cadherin or β-catenin mRNA abundance were observed in fetal tissues between the two groups, proteins encoded by these genes were markedly under-expressed in SCNT trophoblast cells. Immunohistochemistry revealed a different pattern of E-cadherin and total β-catenin localisation in SCNT placentas compared with controls. No difference was observed in subcellular localisation of dephosphorylated active-β-catenin protein in SCNT tissues compared with controls. However, qRT-PCR confirmed that the wingless (WNT)/β-catenin signalling pathway target genes CCND1, CLDN1 and MSX1 were downregulated in SCNT placentas. No differences were detected between two groups of bovine trophectoderm cell lines. Our results suggest that impaired expression of E-cadherin and β-catenin proteins, along with defective β-catenin signalling during embryo attachment, specifically during placentation, is a molecular mechanism explaining insufficient placentation in the bovine SCNT-derived fetus.
Collapse
Affiliation(s)
- H R Kohan-Ghadr
- Department of Clinical Sciences, University of Montreal, 3200 Sicotte, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | | | | | | | | |
Collapse
|
43
|
Dady A, Blavet C, Duband JL. Timing and kinetics of E- to N-cadherin switch during neurulation in the avian embryo. Dev Dyn 2012; 241:1333-49. [PMID: 22684994 DOI: 10.1002/dvdy.23813] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2012] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND During embryonic development, cadherin switches are correlated with tissue remodelings, such as epithelium-to-mesenchyme transition (EMT). An E- to N-cadherin switch also occurs during neurogenesis, but this is not accompanied with EMT. The biological significance of this switch is currently unknown. RESULTS We analyzed the timing and kinetics of the E- to N-cadherin switch during early neural induction and neurulation in the chick embryo, in relation to the patterns of their transcriptional regulators. We found that deployment of the E- to N-cadherin switch program varies considerably along the embryonic axis. Rostrally in regions of primary neurulation, it occurs progressively both in time and space in a manner that appears neither in connection with morphological transformation of neural epithelial cells nor in synchrony with movements of neurulation. Caudally, in regions of secondary neurulation, neurogenesis was not associated with cadherin switch as N-cadherin pre-existed before formation of the neural tube. We also found that, during neural development, cadherin switch is orchestrated by a set of transcriptional regulators distinct from those involved in EMT. CONCLUSIONS Our results indicate that cadherin switch correlates with the partition of the neurectoderm into its three main populations: ectoderm, neural crest, and neural tube.
Collapse
Affiliation(s)
- Alwyn Dady
- Université Pierre et Marie Curie-Paris 6, Laboratoire de Biologie du Développement, Paris, France
| | | | | |
Collapse
|
44
|
Messerschmidt DM, de Vries W, Ito M, Solter D, Ferguson-Smith A, Knowles BB. Trim28 is required for epigenetic stability during mouse oocyte to embryo transition. Science 2012; 335:1499-502. [PMID: 22442485 DOI: 10.1126/science.1216154] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Phenotypic variability in genetic disease is usually attributed to genetic background variation or environmental influence. Here, we show that deletion of a single gene, Trim28 (Kap1 or Tif1β), from the maternal germ line alone, on an otherwise identical genetic background, results in severe phenotypic and epigenetic variability that leads to embryonic lethality. We identify early and minute epigenetic variations in blastomeres of the preimplantation embryo of these animals, suggesting that the embryonic lethality may result from the misregulation of genomic imprinting in mice lacking maternal Trim28. Our results reveal the long-range effects of a maternal gene deletion on epigenetic memory and illustrate the delicate equilibrium of maternal and zygotic factors during nuclear reprogramming.
Collapse
|
45
|
Bedzhov I, Liszewska E, Kanzler B, Stemmler MP. Igf1r signaling is indispensable for preimplantation development and is activated via a novel function of E-cadherin. PLoS Genet 2012; 8:e1002609. [PMID: 22479204 PMCID: PMC3315466 DOI: 10.1371/journal.pgen.1002609] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/05/2012] [Indexed: 01/26/2023] Open
Abstract
Insulin-like growth factor I receptor (Igf1r) signaling controls proliferation, differentiation, growth, and cell survival in many tissues; and its deregulated activity is involved in tumorigenesis. Although important during fetal growth and postnatal life, a function for the Igf pathway during preimplantation development has not been described. We show that abrogating Igf1r signaling with specific inhibitors blocks trophectoderm formation and compromises embryo survival during murine blastocyst formation. In normal embryos total Igf1r is present throughout the membrane, whereas the activated form is found exclusively at cell contact sites, colocalizing with E-cadherin. Using genetic domain switching, we show a requirement for E-cadherin to maintain proper activation of Igf1r. Embryos expressing exclusively a cadherin chimera with N-cadherin extracellular and E-cadherin intracellular domains (NcEc) fail to form a trophectoderm and cells die by apoptosis. In contrast, homozygous mutant embryos expressing a reverse-structured chimera (EcNc) show trophectoderm survival and blastocoel cavitation, indicating a crucial and non-substitutable role of the E-cadherin ectodomain for these processes. Strikingly, blastocyst formation can be rescued in homozygous NcEc embryos by restoring Igf1r signaling, which enhances cell survival. Hence, perturbation of E-cadherin extracellular integrity, independent of its cell-adhesion function, blocked Igf1r signaling and induced cell death in the trophectoderm. Our results reveal an important and yet undiscovered function of Igf1r during preimplantation development mediated by a unique physical interaction between Igf1r and E-cadherin indispensable for proper receptor activation and anti-apoptotic signaling. We provide novel insights into how ligand-dependent Igf1r activity is additionally gated to sense developmental potential in utero and into a bifunctional role of adhesion molecules in contact formation and signaling. One of the most important steps during mammalian development is the formation of a blastocyst before implantation. Proper blastocyst development is fundamentally reliant on the function of the E-cadherin adhesion molecule, which cannot be replaced by another highly related member of the cadherin family. We have addressed the question of how E-cadherin unfolds its unique function during this central embryonic process. We generated mouse mutants that allow specific domain swapping of extra- and intracellular protein domains of E-cadherin with the corresponding portion of N-cadherin. Upon E-cadherin (Cdh1) depletion, apoptosis is induced in cells that are required to form the trophectoderm, the outer cells of a functional blastocyst. Uncoupling of the two E-cadherin domains demonstrated that specifically the presence of the extracellular domain is indispensable in providing essential survival cues. To establish a proper trophectoderm the insulin-like growth factor I receptor (Igf1r) is intimately connected to the E-cadherin–mediated suppression of apoptosis. By interaction of the two proteins Igf1r is efficiently activated to allow embryo survival, blastocyst formation, and implantation. This novel and adhesion-independent function of E-cadherin may serve as paradigm for bifunctionality of adhesion molecules and how they are particularly utilized to interpret signal transduction activities in specific cellular contexts.
Collapse
Affiliation(s)
| | | | | | - Marc P. Stemmler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- * E-mail:
| |
Collapse
|
46
|
Rhee JM, Iannaccone PM. Mapping mouse hemangioblast maturation from headfold stages. Dev Biol 2012; 365:1-13. [PMID: 22426104 DOI: 10.1016/j.ydbio.2012.02.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 11/18/2022]
Abstract
The mouse posterior primitive streak at neural plate/headfold stages (NP/HF, ~7.5 dpc-8 dpc) represents an optimal window from which hemangioblasts can be isolated. We performed immunohistochemistry on this domain using established monoclonal antibodies for proteins that affect blood and endothelial fates. We demonstrate that HoxB4 and GATA1 are the first set of markers that segregate independently to endothelial or blood populations during NP/HF stages of mouse embryonic development. In a subset of cells, both proteins are co-expressed and immunoreactivities appear mutually excluded within nuclear spaces. We searched for this particular state at later sites of hematopoietic stem cell emergence, viz., the aorta-gonad-mesonephros (AGM) and the fetal liver at 10.5-11.5 dpc, and found that only a rare number of cells displayed this character. Based on this spatial-temporal argument, we propose that the earliest blood progenitors emerge either directly from the epiblast or through segregation within the allantoic core domain (ACD) through reduction of cell adhesion and pSmad1/5 nuclear signaling, followed by a stochastic decision toward a blood or endothelial fate that involves GATA1 and HoxB4, respectively. A third form in which binding distributions are balanced may represent a common condition shared by hemangioblasts and HSCs. We developed a heuristic model of hemangioblast maturation, in part, to be explicit about our assumptions.
Collapse
Affiliation(s)
- Jerry M Rhee
- Children's Memorial Research Center, Department of Pediatrics, Developmental Biology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | |
Collapse
|
47
|
Boggetti B, Niessen CM. Adherens junctions in mammalian development, homeostasis and disease: lessons from mice. Subcell Biochem 2012; 60:321-55. [PMID: 22674078 DOI: 10.1007/978-94-007-4186-7_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mice have proven to be a particularly powerful model to study molecular mechanisms of development and disease. The reason for this is the close evolutionary relationship between rodents and humans, similarities in physiological mechanisms in mice and human, and the large number of techniques available to study gene functions in mice. A large number of mice mutations, either germ line, conditional or inducible, have been generated in the past years for adherens junctions components, and the number is still increasing. In this review we will discuss mice models that have contributed to understanding the developmental and physiological role of adherens junctions and their components in mammals and have revealed novel mechanistic aspects of how adherens junctions regulate morphogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Barbara Boggetti
- Department of Dermatology, Center for Molecular Medicine, University of Cologne, Room 4A.05, Robert Kochstrasse 21, 50931, Cologne, Germany
| | | |
Collapse
|
48
|
Straub BK, Rickelt S, Zimbelmann R, Grund C, Kuhn C, Iken M, Ott M, Schirmacher P, Franke WW. E-N-cadherin heterodimers define novel adherens junctions connecting endoderm-derived cells. ACTA ACUST UNITED AC 2011; 195:873-87. [PMID: 22105347 PMCID: PMC3257573 DOI: 10.1083/jcb.201106023] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intercellular junctions play a pivotal role in tissue development and function and also in tumorigenesis. In epithelial cells, decrease or loss of E-cadherin, the hallmark molecule of adherens junctions (AJs), and increase of N-cadherin are widely thought to promote carcinoma progression and metastasis. In this paper, we show that this "cadherin switch" hypothesis does not hold for diverse endoderm-derived cells and cells of tumors derived from them. We show that the cadherins in a major portion of AJs in these cells can be chemically cross-linked in E-N heterodimers. We also show that cells possessing E-N heterodimer AJs can form semistable hemihomotypic AJs with purely N-cadherin-based AJs of mesenchymally derived cells, including stroma cells. We conclude that these heterodimers are the major AJ constituents of several endoderm-derived tissues and tumors and that the prevailing concept of antagonistic roles of these two cadherins in developmental and tumor biology has to be reconsidered.
Collapse
Affiliation(s)
- Beate K Straub
- Helmholtz Group for Cell Biology, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
González S, Ibáñez E, Santaló J. Influence of E-cadherin-mediated cell adhesion on mouse embryonic stem cells derivation from isolated blastomeres. Stem Cell Rev Rep 2011; 7:494-505. [PMID: 21188653 DOI: 10.1007/s12015-010-9221-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Efforts to efficiently derive embryonic stem cells (ESC) from isolated blastomeres have been done to minimize ethical concerns about human embryo destruction. Previous studies in our laboratory indicated a poor derivation efficiency of mouse ESC lines from isolated blastomeres at the 8-cell stage (1/8 blastomeres) due, in part, to a low division rate of the single blastomeres in comparison to their counterparts with a higher number of blastomeres (2/8, 3/8 and 4/8 blastomeres). Communication and adhesion between blastomeres from which the derivation process begins could be important aspects to efficiently derive ESC lines. In the present study, an approach consisting in the adhesion of a chimeric E-cadherin (E-cad-Fc) to the blastomere surface was devised to recreate the signaling produced by native E-cadherin between neighboring blastomeres inside the embryo. By this approach, the division rate of 1/8 blastomeres increased from 44.6% to 88.8% and a short exposure of 24 h to the E-cad-Fc produced an ESC derivation efficiency of 33.6%, significantly higher than the 2.2% obtained from the control group without E-cad-Fc. By contrast, a longer exposure to the same chimeric protein resulted in higher proportions of trophoblastic vesicles. Thus, we establish an important role of E-cadherin-mediated adherens junctions in promoting both the division of single 1/8 blastomeres and the efficiency of the ESC derivation process.
Collapse
Affiliation(s)
- Sheyla González
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| | | | | |
Collapse
|
50
|
Blancas AA, Chen CS, Stolberg S, McCloskey KE. Adhesive forces in embryonic stem cell cultures. Cell Adh Migr 2011; 5:472-9. [PMID: 22274712 PMCID: PMC3277780 DOI: 10.4161/cam.5.6.18270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 09/21/2011] [Accepted: 09/30/2011] [Indexed: 12/19/2022] Open
Abstract
Most cell culture systems grow and spread as contact-inhibited monolayers on flat culture dishes, but the embryonic stem cell (ESC) is one of the cell phenotypes that prefer to self-organize as tightly packed three-dimensional (3D) colonies. ESC also readily form 3D cell aggregates, called embryoid bodies (EB) that partially mimic the spatial and temporal processes of the developing embryo. Here, the rationale for ESC aggregatation, rather than "spreading" on gelatin-coated or mouse embryonic fibroblast (MEF)-coated dishes, is examined through the quantification of the expression levels of adhesion molecules on ESC and the calculation of the adhesive forces on ESC. Modeling each ESC as a dodecahedron, the adhesive force for each ESC-ESC binding was found to be 9.1 x 10(5) pN, whereas, the adhesive force for ESC-MEF binding was found to be an order of magnitude smaller at 7.9 x 10(4) pN. We also show that E-cadherin is the dominating molecule in the ESC-ESC adhesion and blocking E-cadherin leads to a significant reduction in colony formation. Here, we mathematically describe the preference for ESC to self-assemble into ESC-ESC aggregates and 3D colonies, rather than to bind and spread on gelatin or MEF-coated dishes, and have shown that these interactions are predominantly due to E-cadherin expression on ESC.
Collapse
Affiliation(s)
- Alicia A Blancas
- Graduate Program in Quantitative and Systems Biology, University of California Merced, Merced, CA, USA
| | | | | | | |
Collapse
|