1
|
Kim MH, Kuroda M, Ke D, Thanuthanakhun N, Kino-Oka M. An in vitro culture platform for studying the effect of collective cell migration on spatial self-organization within induced pluripotent stem cell colonies. J Biol Eng 2023; 17:25. [PMID: 36998087 PMCID: PMC10064534 DOI: 10.1186/s13036-023-00341-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Human induced pluripotent stem cells (hiPSCs) provide an in vitro system to identify the impact of cell behavior on the earliest stages of cell fate specification during human development. Here, we developed an hiPSC-based model to study the effect of collective cell migration in meso-endodermal lineage segregation and cell fate decisions through the control of space confinement using a detachable ring culture system. RESULTS The actomyosin organization of cells at the edge of undifferentiated colonies formed in a ring barrier differed from that of the cells in the center of the colony. In addition, even in the absence of exogenous supplements, ectoderm, mesoderm, endoderm, and extraembryonic cells differentiated following the induction of collective cell migration at the colony edge by removing the ring-barrier. However, when collective cell migration was inhibited by blocking E-cadherin function, this fate decision within an hiPSC colony was altered to an ectodermal fate. Furthermore, the induction of collective cell migration at the colony edge using an endodermal induction media enhanced endodermal differentiation efficiency in association with cadherin switching, which is involved in the epithelial-mesenchymal transition. CONCLUSIONS Our findings suggest that collective cell migration can be an effective way to drive the segregation of mesoderm and endoderm lineages, and cell fate decisions of hiPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masaki Kuroda
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ding Ke
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Research Base for Cell Manufacturability, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
2
|
Amack JD. Cellular dynamics of EMT: lessons from live in vivo imaging of embryonic development. Cell Commun Signal 2021; 19:79. [PMID: 34294089 PMCID: PMC8296657 DOI: 10.1186/s12964-021-00761-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) refers to a process in which epithelial cells lose apical-basal polarity and loosen cell-cell junctions to take on mesenchymal cell morphologies and invasive properties that facilitate migration through extracellular matrix. EMT-and the reverse mesenchymal-epithelial transition (MET)-are evolutionarily conserved processes that are used throughout embryonic development to drive tissue morphogenesis. During adult life, EMT is activated to close wounds after injury, but also can be used by cancers to promote metastasis. EMT is controlled by several mechanisms that depend on context. In response to cell-cell signaling and/or interactions with the local environment, cells undergoing EMT make rapid changes in kinase and adaptor proteins, adhesion and extracellular matrix molecules, and gene expression. Many of these changes modulate localization, activity, or expression of cytoskeletal proteins that mediate cell shape changes and cell motility. Since cellular changes during EMT are highly dynamic and context-dependent, it is ideal to analyze this process in situ in living organisms. Embryonic development of model organisms is amenable to live time-lapse microscopy, which provides an opportunity to watch EMT as it happens. Here, with a focus on functions of the actin cytoskeleton, I review recent examples of how live in vivo imaging of embryonic development has led to new insights into mechanisms of EMT. At the same time, I highlight specific developmental processes in model embryos-gastrulation in fly and mouse embryos, and neural crest cell development in zebrafish and frog embryos-that provide in vivo platforms for visualizing cellular dynamics during EMT. In addition, I introduce Kupffer's vesicle in the zebrafish embryo as a new model system to investigate EMT and MET. I discuss how these systems have provided insights into the dynamics of adherens junction remodeling, planar cell polarity signaling, cadherin functions, and cytoskeletal organization during EMT, which are not only important for understanding development, but also cancer progression. These findings shed light on mechanisms of actin cytoskeletal dynamics during EMT, and feature live in vivo imaging strategies that can be exploited in future work to identify new mechanisms of EMT and MET. Video Abstract.
Collapse
Affiliation(s)
- Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA. .,BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, USA.
| |
Collapse
|
3
|
Abstract
AbstractAn important goal in the fight against cancer is to understand how tumors become invasive and metastatic. A crucial early step in metastasis is thought to be the epithelial mesenchymal transition (EMT), the process in which epithelial cells transition into a more migratory and invasive, mesenchymal state. Since the genetic regulatory networks driving EMT in tumors derive from those used in development, analysis of EMTs in genetic model organisms such as the vinegar fly, Drosophila melanogaster, can provide great insight into cancer. In this review I highlight the many ways in which studies in the fly are shedding light on cancer metastasis. The review covers both normal developmental events in which epithelial cells become migratory, as well as induced events, whereby normal epithelial cells become metastatic due to genetic manipulations. The ability to make such precise genetic perturbations in the context of a normal, in vivo environment, complete with a working innate immune system, is making the fly increasingly important in understanding metastasis.
Collapse
Affiliation(s)
- Michael J. Murray
- School of BioSciences, Faculty of Science, University of Melbourne, Victoria 3010, Melbourne, Australia
| |
Collapse
|
4
|
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
|
5
|
Liu X, Fan Z, Li Y, Li Z, Zhou Z, Yu X, Wan J, Min Z, Yang L, Li D. microRNA-196a-5p inhibits testicular germ cell tumor progression via NR6A1/E-cadherin axis. Cancer Med 2020; 9:9107-9122. [PMID: 33034957 PMCID: PMC7724306 DOI: 10.1002/cam4.3498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/26/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) are a diverse group of neoplasms that are derived from dysfunctional fetal germ cells and can also present in extragonadal sites. The genetic drivers underlying malignant transformation of TGCTs have not been fully elucidated so far. The aim of the present study is to clarify the functional role and regulatory mechanism of miR‐196a‐5p in TGCTs. We demonstrated that miR‐196a‐5p was downregulated in TGCTs. It can inhibit the proliferation, migration, and invasion of testicular tumor cell lines including NT‐2 and NCCIT through targeting the NR6A1 gene, which we proved its role in promotion of cell proliferation and repression of cellular junction and aggregation. Mechanistically, NR6A1 inhibited E‐cadherin through binding with DR0 sites in the CDH1 gene promoter and recruiting methyltransferases Dnmt1. Further, NR6A1 promoted neuronal marker protein MAP2 expression in RA‐induced neurodifferentiation of NT‐2 cells and testicular tumor xenografts. Clinical histopathologically, NR6A1 was positively correlated with MAP2, and negatively correlated with E‐cadherin in TGCTs. These findings revealed that the miR‐196a‐5p represses cell proliferation, migration, invasion, and tumor neurogenesis by inhibition of NR6A1/E‐cadherin signaling axis, which may be a potential target for diagnosis and therapy of TGCTs.
Collapse
Affiliation(s)
- Xiaowen Liu
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| | - Ziling Fan
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| | - Ye Li
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| | - Zhilan Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China.,Department of Pathology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Zhuan Zhou
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Xuehui Yu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Jingyu Wan
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| | - Ziqian Min
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| | - Lifang Yang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Dan Li
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, P.R. China
| |
Collapse
|
6
|
Massri AJ, Schiebinger GR, Berrio A, Wang L, Wray GA, McClay DR. Methodologies for Following EMT In Vivo at Single Cell Resolution. Methods Mol Biol 2020; 2179:303-314. [PMID: 32939729 DOI: 10.1007/978-1-0716-0779-4_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
An epithelial-mesenchymal transition (EMT) occurs in almost every metazoan embryo at the time mesoderm begins to differentiate. Several embryos have a long record as models for studying an EMT given that a known population of cells enters the EMT at a known time thereby enabling a detailed study of the process. Often, however, it is difficult to learn the molecular details of these model EMT systems because the transitioning cells are a minority of the population of cells in the embryo and in most cases there is an inability to isolate that population. Here we provide a method that enables an examination of genes expressed before, during, and after the EMT with a focus on just the cells that undergo the transition. Single cell RNA-seq (scRNA-seq) has advanced as a technology making it feasible to study the trajectory of gene expression specifically in the cells of interest, in vivo, and without the background noise of other cell populations. The sea urchin skeletogenic cells constitute only 5% of the total number of cells in the embryo yet with scRNA-seq it is possible to study the genes expressed by these cells without background noise. This approach, though not perfect, adds a new tool for uncovering the mechanism of EMT in this cell type.
Collapse
Affiliation(s)
| | | | | | - Lingyu Wang
- Department of Biology, Duke University, Durham, NC, USA
| | | | - David R McClay
- Department of Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
7
|
Cell culture dimensionality influences mesenchymal stem cell fate through cadherin-2 and cadherin-11. Biomaterials 2020; 254:120127. [DOI: 10.1016/j.biomaterials.2020.120127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
|
8
|
Salinas-Saavedra M, Martindale MQ. Par protein localization during the early development of Mnemiopsis leidyi suggests different modes of epithelial organization in the metazoa. eLife 2020; 9:54927. [PMID: 32716297 PMCID: PMC7441587 DOI: 10.7554/elife.54927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
In bilaterians and cnidarians, epithelial cell-polarity is regulated by the interactions between Par proteins, Wnt/PCP signaling pathway, and cell-cell adhesion. Par proteins are highly conserved across Metazoa, including ctenophores. But strikingly, ctenophore genomes lack components of the Wnt/PCP pathway and cell-cell adhesion complexes raising the question if ctenophore cells are polarized by mechanisms involving Par proteins. Here, by using immunohistochemistry and live-cell imaging of specific mRNAs, we describe for the first time the subcellular localization of selected Par proteins in blastomeres and epithelial cells during the embryogenesis of the ctenophore Mnemiopsis leidyi. We show that these proteins distribute differently compared to what has been described for other animals, even though they segregate in a host-specific fashion when expressed in cnidarian embryos. This differential localization might be related to the emergence of different junctional complexes during metazoan evolution.
Collapse
Affiliation(s)
- Miguel Salinas-Saavedra
- The Whitney Laboratory for Marine Bioscience, and the Department of Biology, University of Florida, St. Augustine, United States
| | - Mark Q Martindale
- The Whitney Laboratory for Marine Bioscience, and the Department of Biology, University of Florida, St. Augustine, United States
| |
Collapse
|
9
|
Sun J, Macabenta F, Akos Z, Stathopoulos A. Collective Migrations of Drosophila Embryonic Trunk and Caudal Mesoderm-Derived Muscle Precursor Cells. Genetics 2020; 215:297-322. [PMID: 32487692 PMCID: PMC7268997 DOI: 10.1534/genetics.120.303258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/17/2020] [Indexed: 01/06/2023] Open
Abstract
Mesoderm migration in the Drosophila embryo is a highly conserved, complex process that is required for the formation of specialized tissues and organs, including the somatic and visceral musculature. In this FlyBook chapter, we will compare and contrast the specification and migration of cells originating from the trunk and caudal mesoderm. Both cell types engage in collective migrations that enable cells to achieve new positions within developing embryos and form distinct tissues. To start, we will discuss specification and early morphogenetic movements of the presumptive mesoderm, then focus on the coordinate movements of the two subtypes trunk mesoderm and caudal visceral mesoderm, ending with a comparison of these processes including general insights gained through study.
Collapse
Affiliation(s)
- Jingjing Sun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Frank Macabenta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Zsuzsa Akos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Angelike Stathopoulos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
10
|
The cellular and molecular mechanisms that establish the mechanics of Drosophila gastrulation. Curr Top Dev Biol 2020; 136:141-165. [DOI: 10.1016/bs.ctdb.2019.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
11
|
Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019; 20:69-84. [PMID: 30459476 DOI: 10.1038/s41580-018-0080-4] [Citation(s) in RCA: 2446] [Impact Index Per Article: 407.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular programme that is known to be crucial for embryogenesis, wound healing and malignant progression. During EMT, cell-cell and cell-extracellular matrix interactions are remodelled, which leads to the detachment of epithelial cells from each other and the underlying basement membrane, and a new transcriptional programme is activated to promote the mesenchymal fate. In the context of neoplasias, EMT confers on cancer cells increased tumour-initiating and metastatic potential and a greater resistance to elimination by several therapeutic regimens. In this Review, we discuss recent findings on the mechanisms and roles of EMT in normal and neoplastic tissues, and the cell-intrinsic signals that sustain expression of this programme. We also highlight how EMT gives rise to a variety of intermediate cell states between the epithelial and the mesenchymal state, which could function as cancer stem cells. In addition, we describe the contributions of the tumour microenvironment in inducing EMT and the effects of EMT on the immunobiology of carcinomas.
Collapse
Affiliation(s)
- Anushka Dongre
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA. .,MIT Ludwig Center for Molecular Oncology, Cambridge, MA, USA. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
12
|
Francou A, Anderson KV. The Epithelial-to-Mesenchymal Transition (EMT) in Development and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2019; 4:197-220. [PMID: 34113749 DOI: 10.1146/annurev-cancerbio-030518-055425] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epithelial-to-mesenchymal transitions (EMTs) are complex cellular processes where cells undergo dramatic changes in signaling, transcriptional programming, and cell shape, while directing the exit of cells from the epithelium and promoting migratory properties of the resulting mesenchyme. EMTs are essential for morphogenesis during development and are also a critical step in cancer progression and metastasis formation. Here we provide an overview of the molecular regulation of the EMT process during embryo development, focusing on chick and mouse gastrulation and neural crest development. We go on to describe how EMT regulators participate in the progression of pancreatic and breast cancer in mouse models, and discuss the parallels with developmental EMTs and how these help to understand cancer EMTs. We also highlight the differences between EMTs in tumor and in development to arrive at a broader view of cancer EMT. We conclude by discussing how further advances in the field will rely on in vivo dynamic imaging of the cellular events of EMT.
Collapse
Affiliation(s)
- Alexandre Francou
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065 USA
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065 USA
| |
Collapse
|
13
|
Endo M, Iwawaki T, Yoshimura H, Ozawa T. Photocleavable Cadherin Inhibits Cell-to-Cell Mechanotransduction by Light. ACS Chem Biol 2019; 14:2206-2214. [PMID: 31503442 DOI: 10.1021/acschembio.9b00460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Precise integration of individual cell behaviors is indispensable for collective tissue morphogenesis and maintenance of tissue integrity. Organized multicellular behavior is achieved via mechanical coupling of individual cellular contractility, mediated by cell adhesion molecules at the cell-cell interface. Conventionally, gene depletion or laser microsurgery has been used for functional analysis of intercellular mechanotransduction. Nevertheless, these methods are insufficient to investigate either the spatiotemporal dynamics or the biomolecular contribution in cell-cell mechanical coupling within collective multicellular behaviors. Herein, we present our effort in adaption of PhoCl for attenuation of cell-to-cell tension transmission mediated by E-cadherin. To release intercellular contractile tension applied on E-cadherin molecules with external light, a genetically encoded photocleavable module called PhoCl was inserted into the intracellular domain of E-cadherin, thereby creating photocleavable cadherin (PC-cadherin). In response to light illumination, the PC-cadherin cleaved into two fragments inside cells, resulting in attenuating mechanotransduction at intercellular junctions in living epithelial cells. Light-induced perturbation of the intercellular tension balance with surrounding cells changed the cell shape in an epithelial cell sheet. The method is expected to enable optical manipulation of force-mediated cell-to-cell communications in various multicellular behaviors, which contributes to a deeper understanding of embryogenesis and oncogenesis.
Collapse
Affiliation(s)
- Mizuki Endo
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Takumi Iwawaki
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Hideaki Yoshimura
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Takeaki Ozawa
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| |
Collapse
|
14
|
Pukhlyakova EA, Kirillova AO, Kraus YA, Zimmermann B, Technau U. A cadherin switch marks germ layer formation in the diploblastic sea anemone Nematostella vectensis. Development 2019; 146:dev.174623. [PMID: 31540916 DOI: 10.1242/dev.174623] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 09/12/2019] [Indexed: 02/01/2023]
Abstract
Morphogenesis is a shape-building process during development of multicellular organisms. During this process, the establishment and modulation of cell-cell contacts play an important role. Cadherins, the major cell adhesion molecules, form adherens junctions connecting epithelial cells. Numerous studies of Bilateria have shown that cadherins are associated with the regulation of cell differentiation, cell shape changes, cell migration and tissue morphogenesis. To date, the role of cadherins in non-bilaterians is unknown. Here, we study the expression and function of two paralogous classical cadherins, Cadherin 1 and Cadherin 3, in a diploblastic animal, the sea anemone Nematostella vectensis We show that a cadherin switch accompanies the formation of germ layers. Using specific antibodies, we show that both cadherins are localized to adherens junctions at apical and basal positions in ectoderm and endoderm. During gastrulation, partial epithelial-to-mesenchymal transition of endodermal cells is marked by stepwise downregulation of Cadherin 3 and upregulation of Cadherin 1. Knockdown experiments show that both cadherins are required for maintenance of tissue integrity and tissue morphogenesis. Thus, both sea anemones and bilaterians use independently duplicated cadherins combinatorially for tissue morphogenesis and germ layer differentiation.
Collapse
Affiliation(s)
- Ekaterina A Pukhlyakova
- Department for Molecular Evolution and Development, Centre of Organismal Systems Biology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Anastasia O Kirillova
- Department for Molecular Evolution and Development, Centre of Organismal Systems Biology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.,Department of Evolutionary Biology, Biological Faculty, Moscow State University, Leninskie Gory 1/12, 119991 Moscow, Russia
| | - Yulia A Kraus
- Department of Evolutionary Biology, Biological Faculty, Moscow State University, Leninskie Gory 1/12, 119991 Moscow, Russia
| | - Bob Zimmermann
- Department for Molecular Evolution and Development, Centre of Organismal Systems Biology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Ulrich Technau
- Department for Molecular Evolution and Development, Centre of Organismal Systems Biology, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| |
Collapse
|
15
|
The Transcription Factor Elf3 Is Essential for a Successful Mesenchymal to Epithelial Transition. Cells 2019; 8:cells8080858. [PMID: 31404945 PMCID: PMC6721682 DOI: 10.3390/cells8080858] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/22/2019] [Accepted: 07/27/2019] [Indexed: 12/13/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) and the mesenchymal to epithelial transition (MET) are two critical biological processes that are involved in both physiological events such as embryogenesis and development and also pathological events such as tumorigenesis. They present with dramatic changes in cellular morphology and gene expression exhibiting acute changes in E-cadherin expression. Despite the comprehensive understanding of EMT, the regulation of MET is far from being understood. To find novel regulators of MET, we hypothesized that such factors would correlate with Cdh1 expression. Bioinformatics examination of several expression profiles suggested Elf3 as a strong candidate. Depletion of Elf3 at the onset of MET severely impaired the progression to the epithelial state. This MET defect was explained, in part, by the absence of E-cadherin at the plasma membrane. Moreover, during MET, ELF3 interacts with the Grhl3 promoter and activates its expression. Our findings present novel insights into the regulation of MET and reveal ELF3 as an indispensable guardian of the epithelial state. A better understanding of MET will, eventually, lead to better management of metastatic cancers.
Collapse
|
16
|
Pütz SM. Mbt/PAK4 together with SRC modulates N-Cadherin adherens junctions in the developing Drosophila eye. Biol Open 2019; 8:8/3/bio038406. [PMID: 30885947 PMCID: PMC6451336 DOI: 10.1242/bio.038406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Tissue morphogenesis is accompanied by changes of adherens junctions (AJ). During Drosophila eye development, AJ reorganization includes the formation of isolated N-Cadherin AJ between photoreceptors R3/R4. Little is known about how these N-Cadherin AJ are established and maintained. This study focuses on the kinases Mbt/PAK4 and SRC, both known to alter E-Cadherin AJ across phyla. Drosophila p21-activated kinase Mbt and the non-receptor tyrosine kinases Src64 and Src42 regulate proper N-Cadherin AJ. N-Cadherin AJ elongation depends on SRC kinase activity. Cell culture experiments demonstrate binding of both Drosophila SRC isoforms to N-Cadherin and its subsequent tyrosine phosphorylation. In contrast, Mbt stabilizes but does not bind N-Cadherin in vitro. Mbt is required in R3/R4 for zipping the N-Cadherin AJ between these cells, independent of its kinase activity and Cdc42-binding. The mbt phenotype can be reverted by mutations in Src64 and Src42. Because Mbt neither directly binds to SRC proteins nor has a reproducible influence on their kinase activity, the conclusion is that Mbt and SRC signaling converge on N-Cadherin. N-Cadherin AJ formation during eye development requires a proper balance between the promoting effects of Mbt and the inhibiting influences of SRC kinases. Summary: N-Cadherin adherens junction formation in the Drosophila larval eye imaginal disc is controlled by the combined functions of the p21-activated kinase Mbt/PAK4 and the kinases Src64 and Src42.
Collapse
Affiliation(s)
- Stephanie M Pütz
- Institute of Medical Radiation and Cell Research, University of Würzburg, Biozentrum, Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|
17
|
Lv S, Wang N, Lv H, Yang J, Liu J, Li WP, Zhang C, Chen ZJ. The Attenuation of Trophoblast Invasion Caused by the Downregulation of EZH2 Is Involved in the Pathogenesis of Human Recurrent Miscarriage. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 14:377-387. [PMID: 30710891 PMCID: PMC6356049 DOI: 10.1016/j.omtn.2018.12.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/28/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022]
Abstract
Recurrent miscarriage (RM) is currently defined as two or more losses of a clinically established intrauterine pregnancy. Despite years of research, RM continues to be a clinically frustrating challenge for patients and physicians, and its etiology remains poorly understood. Accumulating evidence has suggested that epigenetic modifications are involved in early embryogenesis, and defects in epigenetic patterning contribute to the development of RM. Here, we studied the role of enhancer of zeste homolog 2 (EZH2) in the pathogenesis of RM and found that the EZH2 expression was significantly decreased in the villi from women with RM compared with that in control villi. EZH2 promoted the invasion of trophoblast cells. Moreover, EZH2 could promote epithelial-mesenchymal transition by epigenetically silencing CDX1. Both chromatin immunoprecipitation (ChIP)-PCR and dual-luciferase report assays demonstrated that EZH2 repressed CDX1 transcription via direct binding to its promoter region and then trimethylating Histone3-Lysine27. Furthermore, we discovered that progesterone, which is used extensively in the treatment of miscarriage and RM, increased the expression of EZH2 via the extracellular signaling-regulated kinase (ERK1/2) pathway. These findings revealed that EZH2 may regulate trophoblast invasion as an epigenetic factor, suggesting that EZH2 might be a potential therapeutic target for RM.
Collapse
Affiliation(s)
- Shijian Lv
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Na Wang
- Obstetrical Department, Obstetrics and Gynecology Hospital of Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai 200090, China
| | - Hong Lv
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jieqiong Yang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jianwei Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Wei-Ping Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Ji'nan, Shandong 250014, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
18
|
Wang H, Unternaehrer JJ. Epithelial-mesenchymal Transition and Cancer Stem Cells: At the Crossroads of Differentiation and Dedifferentiation. Dev Dyn 2018; 248:10-20. [DOI: 10.1002/dvdy.24678] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/29/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Hanmin Wang
- Division of Biochemistry, Department of Basic Sciences; Loma Linda University; Loma Linda California
| | - Juli J. Unternaehrer
- Division of Biochemistry, Department of Basic Sciences; Loma Linda University; Loma Linda California
| |
Collapse
|
19
|
Sun J, Stathopoulos A. FGF controls epithelial-mesenchymal transitions during gastrulation by regulating cell division and apicobasal polarity. Development 2018; 145:dev.161927. [PMID: 30190277 DOI: 10.1242/dev.161927] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 08/31/2018] [Indexed: 01/06/2023]
Abstract
To support tissue and organ development, cells transition between epithelial and mesenchymal states. Here, we have investigated how mesoderm cells change state in Drosophila embryos and whether fibroblast growth factor (FGF) signaling plays a role. During gastrulation, presumptive mesoderm cells invaginate, undergo an epithelial-to-mesenchymal state transition (EMT) and migrate upon the ectoderm. Our data show that EMT is a prolonged process in which adherens junctions progressively decrease in number throughout the migration of mesoderm cells. FGF influences adherens junction number and promotes mesoderm cell division, which we propose decreases cell-cell attachments to support slow EMT while retaining collective cell movement. We also found that, at the completion of migration, cells form a monolayer and undergo a reverse mesenchymal-to-epithelial transition (MET). FGF activity leads to accumulation of β-integrin Myospheroid basally and cell polarity factor Bazooka apically within mesoderm cells, thereby reestablishing apicobasal cell polarity in an epithelialized state in which cells express both E-Cadherin and N-Cadherin. In summary, FGF plays a dynamic role in supporting mesoderm cell development to ensure collective mesoderm cell movements, as well as proper differentiation of mesoderm cell types.
Collapse
Affiliation(s)
- Jingjing Sun
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| |
Collapse
|
20
|
Rogers CD, Sorrells LK, Bronner ME. A catenin-dependent balance between N-cadherin and E-cadherin controls neuroectodermal cell fate choices. Mech Dev 2018; 152:44-56. [PMID: 30009960 PMCID: PMC6112866 DOI: 10.1016/j.mod.2018.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/16/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023]
Abstract
Characterizing endogenous protein expression, interaction and function, this study identifies in vivo interactions and competitive balance between N-cadherin and E-cadherin in developing avian (Gallus gallus) neural and neural crest cells. Numerous cadherin proteins, including neural cadherin (Ncad) and epithelial cadherin (Ecad), are expressed in the developing neural plate as well as in neural crest cells as they delaminate from the newly closed neural tube. To clarify independent or coordinate function during development, we examined their expression in the cranial region. The results revealed surprising overlap and distinct localization of Ecad and Ncad in the neural tube. Using a proximity ligation assay and co-immunoprecipitation, we found that Ncad and Ecad formed heterotypic complexes in the developing neural tube, and that modulation of Ncad levels led to reciprocal gain or reduction of Ecad protein, which then alters ectodermal cell fate. Here, we demonstrate that the balance of Ecad and Ncad is dependent upon the availability of β-catenin proteins, and that alteration of either classical cadherin modifies the proportions of the neural crest and neuroectodermal cells that are specified.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Lisa K Sorrells
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering 139-74, California Institute of Technology, Pasadena, CA 91125, United States of America.
| |
Collapse
|
21
|
Salinas-Saavedra M, Rock AQ, Martindale MQ. Germ layer-specific regulation of cell polarity and adhesion gives insight into the evolution of mesoderm. eLife 2018; 7:e36740. [PMID: 30063005 PMCID: PMC6067901 DOI: 10.7554/elife.36740] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/29/2018] [Indexed: 12/20/2022] Open
Abstract
In triploblastic animals, Par-proteins regulate cell-polarity and adherens junctions of both ectodermal and endodermal epithelia. But, in embryos of the diploblastic cnidarian Nematostella vectensis, Par-proteins are degraded in all cells in the bifunctional gastrodermal epithelium. Using immunohistochemistry, CRISPR/Cas9 mutagenesis, and mRNA overexpression, we describe the functional association between Par-proteins, ß-catenin, and snail transcription factor genes in N. vectensis embryos. We demonstrate that the aPKC/Par complex regulates the localization of ß-catenin in the ectoderm by stabilizing its role in cell-adhesion, and that endomesodermal epithelial cells are organized by a different cell-adhesion system than overlying ectoderm. We also show that ectopic expression of snail genes, which are expressed in mesodermal derivatives in bilaterians, is sufficient to downregulate Par-proteins and translocate ß-catenin from the junctions to the cytoplasm in ectodermal cells. These data provide molecular insight into the evolution of epithelial structure and distinct cell behaviors in metazoan embryos.
Collapse
Affiliation(s)
- Miguel Salinas-Saavedra
- The Whitney
Laboratory for Marine BioscienceUniversity of
FloridaFloridaUnited
States
- Department of
BiologyUniversity of
FloridaFloridaUnited
States
| | - Amber Q Rock
- The Whitney
Laboratory for Marine BioscienceUniversity of
FloridaFloridaUnited
States
| | - Mark Q Martindale
- The Whitney
Laboratory for Marine BioscienceUniversity of
FloridaFloridaUnited
States
- Department of
BiologyUniversity of
FloridaFloridaUnited
States
| |
Collapse
|
22
|
Campbell K. Contribution of epithelial-mesenchymal transitions to organogenesis and cancer metastasis. Curr Opin Cell Biol 2018; 55:30-35. [PMID: 30006053 PMCID: PMC6284102 DOI: 10.1016/j.ceb.2018.06.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/10/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT) plays crucial roles during development, and inappropriate activation of EMTs are associated with tumor progression and promoting metastasis. In recent years, increasing studies have identified developmental contexts where cells undergo an EMT and transition to a partial-state, downregulating just a subset of epithelial characteristics and increasing only some mesenchymal traits, such as invasive motility. In parallel, recent studies have shown that EMTs are rarely fully activated in tumor cells, generating a diverse array of transition states. As our appreciation of the full spectrum of intermediate phenotypes and the huge diversity in underlying mechanisms grows, cross-disciplinary collaborations investigating developmental-EMTs and cancer-EMTs will be fundamental in order to achieve a full mechanistic understanding of this complex cell process.
Collapse
Affiliation(s)
- Kyra Campbell
- Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK; Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, UK.
| |
Collapse
|
23
|
Cousin H. Cadherins function during the collective cell migration of Xenopus Cranial Neural Crest cells: revisiting the role of E-cadherin. Mech Dev 2017; 148:79-88. [PMID: 28467887 PMCID: PMC5662486 DOI: 10.1016/j.mod.2017.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/21/2017] [Accepted: 04/28/2017] [Indexed: 12/20/2022]
Abstract
Collective cell migration is a process whereby cells move while keeping contact with other cells. The Xenopus Cranial Neural Crest (CNC) is a population of cells that emerge during early embryogenesis and undergo extensive migration from the dorsal to ventral part of the embryo's head. These cells migrate collectively and require cadherin mediated cell-cell contact. In this review, we will describe the key features of Xenopus CNC migration including the key molecules driving their migration. We will also review the role of the various cadherins during Xenopus CNC emergence and migration. Lastly, we will discuss the recent and seemingly controversial findings showing that E-cadherin presence is essential for CNC migration.
Collapse
Affiliation(s)
- Hélène Cousin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
24
|
Giger FA, David NB. Endodermal germ-layer formation through active actin-driven migration triggered by N-cadherin. Proc Natl Acad Sci U S A 2017; 114:10143-10148. [PMID: 28874564 PMCID: PMC5617292 DOI: 10.1073/pnas.1708116114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Germ-layer formation during gastrulation is both a fundamental step of development and a paradigm for tissue formation and remodeling. However, the cellular and molecular basis of germ-layer segregation is poorly understood, mostly because of the lack of direct in vivo observations. We used mosaic zebrafish embryos to investigate the formation of the endoderm. High-resolution live imaging and functional analyses revealed that endodermal cells reach their characteristic innermost position through an active, oriented, and actin-based migration dependent on Rac1, which contrasts with the previously proposed differential adhesion cell sorting. Rather than being attracted to their destination, the yolk syncytial layer, cells appear to migrate away from their neighbors. This migration depends on N-cadherin that, when imposed in ectodermal cells, is sufficient to trigger their internalization without affecting their fate. Overall, these results lead to a model of germ-layer formation in which, upon N-cadherin expression, endodermal cells actively migrate away from their epiblastic neighbors to reach their internal position, revealing cell-contact avoidance as an unexplored mechanism driving germ-layer formation.
Collapse
Affiliation(s)
- Florence A Giger
- CNRS UMR8197, F-75005 Paris, France
- INSERM U1024, F-75005 Paris, France
- Institut de Biologie de l'Ecole Normale Supérieure, F-75005 Paris, France
| | - Nicolas B David
- CNRS UMR8197, F-75005 Paris, France;
- INSERM U1024, F-75005 Paris, France
- Institut de Biologie de l'Ecole Normale Supérieure, F-75005 Paris, France
- Laboratory for Optics and Biosciences, Ecole Polytechnique, 91128 Palaiseau, France
| |
Collapse
|
25
|
Acloque H, Ocaña OH, Abad D, Stern CD, Nieto MA. Snail2 and Zeb2 repress P-cadherin to define embryonic territories in the chick embryo. Development 2017; 144:649-656. [PMID: 28087626 DOI: 10.1242/dev.142562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/23/2016] [Indexed: 12/12/2022]
Abstract
Snail and Zeb transcription factors induce epithelial-to-mesenchymal transition (EMT) in embryonic and adult tissues by direct repression of E-cadherin transcription. The repression of E-cadherin transcription by the EMT inducers Snail1 and Zeb2 plays a fundamental role in defining embryonic territories in the mouse, as E-cadherin needs to be downregulated in the primitive streak and in the epiblast, concomitant with the formation of mesendodermal precursors and the neural plate, respectively. Here, we show that in the chick embryo, E-cadherin is weakly expressed in the epiblast at pre-primitive streak stages where it is substituted for by P-cadherin We also show that Snail2 and Zeb2 repress P-cadherin transcription in the primitive streak and the neural plate, respectively. This indicates that E- and P-cadherin expression patterns evolved differently between chick and mouse. As such, the Snail1/E-cadherin axis described in the early mouse embryo corresponds to Snail2/P-cadherin in the chick, but both Snail factors and Zeb2 fulfil a similar role in chick and mouse in directly repressing ectodermal cadherin genes to contribute to the delamination of mesendodermal precursors at gastrulation and the proper specification of the neural ectoderm during neural induction.
Collapse
Affiliation(s)
- Hervé Acloque
- Instituto de Neurociencias de Alicante, CSIC-UMH, Avenida Ramón y Cajal s/n, San Juan de Alicante 03550, Spain .,GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan 31326, France
| | - Oscar H Ocaña
- Instituto de Neurociencias de Alicante, CSIC-UMH, Avenida Ramón y Cajal s/n, San Juan de Alicante 03550, Spain
| | - Diana Abad
- Instituto de Neurociencias de Alicante, CSIC-UMH, Avenida Ramón y Cajal s/n, San Juan de Alicante 03550, Spain
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - M Angela Nieto
- Instituto de Neurociencias de Alicante, CSIC-UMH, Avenida Ramón y Cajal s/n, San Juan de Alicante 03550, Spain
| |
Collapse
|
26
|
Abstract
The significant parallels between cell plasticity during embryonic development and carcinoma progression have helped us understand the importance of the epithelial-mesenchymal transition (EMT) in human disease. Our expanding knowledge of EMT has led to a clarification of the EMT program as a set of multiple and dynamic transitional states between the epithelial and mesenchymal phenotypes, as opposed to a process involving a single binary decision. EMT and its intermediate states have recently been identified as crucial drivers of organ fibrosis and tumor progression, although there is some need for caution when interpreting its contribution to metastatic colonization. Here, we discuss the current state-of-the-art and latest findings regarding the concept of cellular plasticity and heterogeneity in EMT. We raise some of the questions pending and identify the challenges faced in this fast-moving field.
Collapse
|
27
|
Weng M, Wieschaus E. Polarity protein Par3/Bazooka follows myosin-dependent junction repositioning. Dev Biol 2017; 422:125-134. [PMID: 28063874 DOI: 10.1016/j.ydbio.2017.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 11/24/2022]
Abstract
The polarity protein Par3/Bazooka (Baz) has been established as a central component of the apical basal polarity system that determines the position of cell-cell junctions in epithelial cells. Consistent with that view, we show that shortly before gastrulation in Drosophila, Baz protein in the mesoderm is down-regulated from junctional sites in response to Snail (Sna) expression. This down-regulation leads to a specific decrease in adherens junctions without affecting other E-Cadherin pools. However, we further show that, interactions between Baz and junctions are not unidirectional. During apical constriction and the internalization of the mesoderm, down-regulation of Baz is transiently blocked as adherens junctions shift apically and are strengthened in response to tension generated by contractile actomyosin. When such junction remodeling is prevented by down-regulating myosin, Baz is lost prematurely in mesodermal epithelium. During such apical shifts, Baz is initially left behind as the junction shifts position, but then re-accumulates at the new location of the junctions. On the dorsal side of the embryo, a similar pattern of myosin activity appears to limit the basal shift in junctions normally driven by Baz that controls epithelium folding. Our results suggest a model where the sensitivity of Baz to Sna expression leads to the Sna-dependent junction disassembly required for a complete epithelium-mesenchymal transition. Meanwhile this loss of Baz-dependent junction maintenance is countered by the myosin-based mechanism which promotes an apical shift and strengthening of junctions accompanied by a transient re-positioning and maintenance of Baz proteins.
Collapse
Affiliation(s)
- Mo Weng
- Department of Molecular Biology, Princeton University, USA
| | - Eric Wieschaus
- Department of Molecular Biology, Princeton University, USA; Howard Hughes Medical Institute, Princeton University, Princeton, NJ 08540, USA.
| |
Collapse
|
28
|
Campbell K, Casanova J. A common framework for EMT and collective cell migration. Development 2016; 143:4291-4300. [DOI: 10.1242/dev.139071] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During development, cells often switch between static and migratory behaviours. Such transitions are fundamental events in development and are linked to harmful consequences in pathology. It has long been considered that epithelial cells either migrate collectively as epithelial cells, or undergo an epithelial-to-mesenchymal transition and migrate as individual mesenchymal cells. Here, we assess what is currently known about in vivo cell migratory phenomena and hypothesise that such migratory behaviours do not fit into alternative and mutually exclusive categories. Rather, we propose that these categories can be viewed as the most extreme cases of a general continuum of morphological variety, with cells harbouring different degrees or combinations of epithelial and mesenchymal features and displaying an array of migratory behaviours.
Collapse
Affiliation(s)
- Kyra Campbell
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| |
Collapse
|
29
|
Stollewerk A. A flexible genetic toolkit for arthropod neurogenesis. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150044. [PMID: 26598727 DOI: 10.1098/rstb.2015.0044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Arthropods show considerable variations in early neurogenesis. This includes the pattern of specification, division and movement of neural precursors and progenitors. In all metazoans with nervous systems, including arthropods, conserved genes regulate neurogenesis, which raises the question of how the various morphological mechanisms have emerged and how the same genetic toolkit might generate different morphological outcomes. Here I address this question by comparing neurogenesis across arthropods and show how variations in the regulation and function of the neural genes might explain this phenomenon and how they might have facilitated the evolution of the diverse morphological mechanisms of neurogenesis.
Collapse
Affiliation(s)
- Angelika Stollewerk
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
30
|
Alaee M, Danesh G, Pasdar M. Plakoglobin Reduces the in vitro Growth, Migration and Invasion of Ovarian Cancer Cells Expressing N-Cadherin and Mutant p53. PLoS One 2016; 11:e0154323. [PMID: 27144941 PMCID: PMC4856367 DOI: 10.1371/journal.pone.0154323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/12/2016] [Indexed: 12/20/2022] Open
Abstract
Aberrant expression of cadherins and catenins plays pivotal roles in ovarian cancer development and progression. Plakoglobin (PG, γ-catenin) is a paralog of β-catenin with dual adhesive and signaling functions. While β-catenin has known oncogenic function, PG generally acts as a tumor/metastasis suppressor. We recently showed that PG interacted with p53 and that its growth/metastasis inhibitory function may be mediated by this interaction. Very little is known about the role of PG in ovarian cancer. Here, we investigated the in vitro tumor/metastasis suppressor effects of PG in ovarian cancer cell lines with mutant p53 expression and different cadherin profiles. We showed that the N-cadherin expressing and E-cadherin and PG deficient ES-2 cells were highly migratory and invasive, whereas OV-90 cells that express E-cadherin, PG and very little/no N-cadherin were not. Exogenous expression of PG or E-cadherin or N-cadherin knockdown in ES-2 cells (ES-2-E-cad, ES-2-PG and ES-2-shN-cad) significantly reduced their migration and invasion. Also, PG expression or N-cadherin knockdown significantly decreased ES-2 cells growth. Furthermore, PG interacted with both cadherins and with wild type and mutant p53 in normal ovarian and ES-2-PG cell lines, respectively.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Ghazal Danesh
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
- * E-mail:
| |
Collapse
|
31
|
Moly PK, Cooley JR, Zeltzer SL, Yatskievych TA, Antin PB. Gastrulation EMT Is Independent of P-Cadherin Downregulation. PLoS One 2016; 11:e0153591. [PMID: 27097030 PMCID: PMC4838233 DOI: 10.1371/journal.pone.0153591] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 03/31/2016] [Indexed: 11/24/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an evolutionarily conserved process during which cells lose epithelial characteristics and gain a migratory phenotype. Although downregulation of epithelial cadherins by Snail and other transcriptional repressors is generally considered a prerequisite for EMT, recent studies have challenged this view. Here we investigate the relationship between E-cadherin and P-cadherin expression and localization, Snail function and EMT during gastrulation in chicken embryos. Expression analyses show that while E-cadherin transcripts are detected in the epiblast but not in the primitive streak or mesoderm, P-cadherin mRNA and protein are present in the epiblast, primitive and mesoderm. Antibodies that specifically recognize E-cadherin are not presently available. During EMT, P-cadherin relocalizes from the lateral surfaces of epithelial epiblast cells to a circumferential distribution in emerging mesodermal cells. Cells electroporated with an E-cadherin expression construct undergo EMT and migrate into the mesoderm. An examination of Snail function showed that reduction of Slug (SNAI2) protein levels using a morpholino fails to inhibit EMT, and expression of human or chicken Snail in epiblast cells fails to induce EMT. In contrast, cells expressing the Rho inhibitor peptide C3 rapidly exit the epiblast without activating Slug or the mesoderm marker N-cadherin. Together, these experiments show that epiblast cells undergo EMT while retaining P-cadherin, and raise questions about the mechanisms of EMT regulation during avian gastrulation.
Collapse
Affiliation(s)
- Pricila K. Moly
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - James R. Cooley
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Sebastian L. Zeltzer
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Tatiana A. Yatskievych
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Parker B. Antin
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
- * E-mail:
| |
Collapse
|
32
|
Precise long-range migration results from short-range stepwise migration during ring gland organogenesis. Dev Biol 2016; 414:45-57. [PMID: 27063193 DOI: 10.1016/j.ydbio.2016.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/04/2016] [Accepted: 04/04/2016] [Indexed: 01/21/2023]
Abstract
Many organs are specified far from the location they occupy when functional, having to migrate long distances through the heterogeneous and dynamic environment of the early embryo. We study the formation of the main Drosophila endocrine organ, the ring gland, as a new model to investigate in vivo the genetic regulation of collective cell migration. The ring gland results from the fusion of three independent gland primordia that migrate from the head towards the anterior aorta as the embryo is experiencing major morphogenetic movements. To complete their long-range migration, the glands extend filopodia moving sequentially towards a nearby intermediate target and from there to more distal ones. Thus, the apparent long-range migration is composed of several short-range migratory steps that facilitate reaching the final destination. We find that the target tissues react to the gland's proximity by sending filopodia towards it. Our finding of a succession of independent migration stages is consistent with the stepwise evolution of ring gland assembly and fits with the observed gland locations found in extant crustaceans, basal insects and flies.
Collapse
|
33
|
Weng M, Wieschaus E. Myosin-dependent remodeling of adherens junctions protects junctions from Snail-dependent disassembly. J Cell Biol 2016; 212:219-29. [PMID: 26754645 PMCID: PMC4738385 DOI: 10.1083/jcb.201508056] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/10/2015] [Indexed: 11/22/2022] Open
Abstract
Although Snail is essential for disassembly of adherens junctions during epithelial-mesenchymal transitions (EMTs), loss of adherens junctions in Drosophila melanogaster gastrula is delayed until mesoderm is internalized, despite the early expression of Snail in that primordium. By combining live imaging and quantitative image analysis, we track the behavior of E-cadherin-rich junction clusters, demonstrating that in the early stages of gastrulation most subapical clusters in mesoderm not only persist, but move apically and enhance in density and total intensity. All three phenomena depend on myosin II and are temporally correlated with the pulses of actomyosin accumulation that drive initial cell shape changes during gastrulation. When contractile myosin is absent, the normal Snail expression in mesoderm, or ectopic Snail expression in ectoderm, is sufficient to drive early disassembly of junctions. In both cases, junctional disassembly can be blocked by simultaneous induction of myosin contractility. Our findings provide in vivo evidence for mechanosensitivity of cell-cell junctions and imply that myosin-mediated tension can prevent Snail-driven EMT.
Collapse
Affiliation(s)
- Mo Weng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540 Howard Hughes Medical Institute, Princeton University, Princeton, NJ 08540
| | - Eric Wieschaus
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540 Howard Hughes Medical Institute, Princeton University, Princeton, NJ 08540
| |
Collapse
|
34
|
Pocha SM, Montell DJ. Cellular and molecular mechanisms of single and collective cell migrations in Drosophila: themes and variations. Annu Rev Genet 2015; 48:295-318. [PMID: 25421599 DOI: 10.1146/annurev-genet-120213-092218] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The process of cell migration is essential throughout life, driving embryonic morphogenesis and ensuring homeostasis in adults. Defects in cell migration are a major cause of human disease, with excessive migration causing autoimmune diseases and cancer metastasis, whereas reduced capacity for migration leads to birth defects and immunodeficiencies. Myriad studies in vitro have established a consensus view that cell migrations require cell polarization, Rho GTPase-mediated cytoskeletal rearrangements, and myosin-mediated contractility. However, in vivo studies later revealed a more complex picture, including the discovery that cells migrate not only as single units but also as clusters, strands, and sheets. In particular, the role of E-Cadherin in cell motility appears to be more complex than previously appreciated. Here, we discuss recent advances achieved by combining the plethora of genetic tools available to the Drosophila geneticist with live imaging and biophysical techniques. Finally, we discuss the emerging themes such studies have revealed and ponder the puzzles that remain to be solved.
Collapse
Affiliation(s)
- Shirin M Pocha
- Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California; 93106-9625; ,
| | | |
Collapse
|
35
|
Maartens AP, Brown NH. The many faces of cell adhesion during Drosophila muscle development. Dev Biol 2015; 401:62-74. [DOI: 10.1016/j.ydbio.2014.12.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
|
36
|
Fagotto F. Regulation of Cell Adhesion and Cell Sorting at Embryonic Boundaries. Curr Top Dev Biol 2015; 112:19-64. [DOI: 10.1016/bs.ctdb.2014.11.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Park KS, Dubon MJ, Gumbiner BM. N-cadherin mediates the migration of MCF-10A cells undergoing bone morphogenetic protein 4-mediated epithelial mesenchymal transition. Tumour Biol 2014; 36:3549-56. [PMID: 25542234 DOI: 10.1007/s13277-014-2991-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/17/2014] [Indexed: 01/08/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) of mammary epithelial cells is important in both normal morphogenesis of mammary glands and metastasis of breast cancer. Cadherin switching from E-cadherin to N-cadherin plays important roles in EMT. We found that cadherin switching is important in bone morphogenetic protein 4 (BMP4)-induced EMT in MCF-10A cells. BMP4 increased the phosphorylation of SMAD proteins in MCF-10A cells. Canonical BMP4 signaling decreased the expression of E-cadherin and disrupted the polarity of the tight junction protein ZO-1 in MCF-10A cells. However, the expression of N-cadherin and SNAI2 was up-regulated in BMP4-treated MCF-10A cells. MCF-10A cells that expressed N-cadherin migrated into type I collagen gels in response to BMP4 when evaluated using three-dimensional culture assays. Thus, active canonical BMP4 signaling is important for the migration and EMT of mammary epithelial cells. Moreover, the decrease in E-cadherin and/or increase in N-cadherin may be required for BMP4-induced migration and EMT.
Collapse
Affiliation(s)
- Ki-Sook Park
- East-West Medical Research Institute/College of Medicine, Kyung Hee University, Seoul, 130-701, Korea,
| | | | | |
Collapse
|
38
|
Ectopic expression screen identifies genes affecting Drosophila mesoderm development including the HSPG Trol. G3-GENES GENOMES GENETICS 2014; 5:301-13. [PMID: 25538103 PMCID: PMC4321038 DOI: 10.1534/g3.114.015891] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Gastrulation of the embryo involves coordinate cell movements likely supported by multiple signaling pathways, adhesion molecules, and extracellular matrix components. Fibroblast growth factors (FGFs) have a major role in Drosophila melanogaster mesoderm migration; however, few other inputs are known and the mechanism supporting cell movement is unclear. To provide insight, we performed an ectopic expression screen to identify secreted or membrane-associated molecules that act to support mesoderm migration. Twenty-four UAS insertions were identified that cause lethality when expressed in either the mesoderm (Twi-Gal4) or the ectoderm (69B-Gal4). The list was narrowed to a subset of 10 genes that were shown to exhibit loss-of-function mutant phenotypes specifically affecting mesoderm migration. These include the FGF ligand Pyramus, α-integrins, E-cadherin, Cueball, EGFR, JAK/STAT signaling components, as well as the heparan sulfate proteoglycan (HSPG) Terribly reduced optic lobes (Trol). Trol encodes the ortholog of mammalian HSPG Perlecan, a demonstrated FGF signaling cofactor. Here, we examine the role of Trol in Drosophila mesoderm migration and compare and contrast its role with that of Syndecan (Sdc), another HSPG previously implicated in this process. Embryos mutant for Trol or Sdc were obtained and analyzed. Our data support the view that both HSPGs function to support FGF-dependent processes in the early embryo as they share phenotypes with FGF mutants: Trol in terms of effects on mesoderm migration and caudal visceral mesoderm (CVM) migration and Sdc in terms of dorsal mesoderm specification. The differential roles uncovered for these two HSPGs suggest that HSPG cofactor choice may modify FGF-signaling outputs.
Collapse
|
39
|
Qi L, Sun B, Liu Z, Cheng R, Li Y, Zhao X. Wnt3a expression is associated with epithelial-mesenchymal transition and promotes colon cancer progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:107. [PMID: 25499541 PMCID: PMC4269867 DOI: 10.1186/s13046-014-0107-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
Introduction Epithelial–mesenchymal transition (EMT) contributes to the progression and metastasis of cancer cells and is associated with a more invasive phenotype of cancer. The Wnt/β-catenin signaling pathway is one of the major pathways involved in EMT regulation. Many studies provide evidence that β-catenin, the key regulator of the canonical Wnt signaling pathway, is important in regulating EMT in cancer. However, the roles of Wnt3a, the representative canonical Wnt ligand, in EMT and colon cancer progression have not yet been fully explored. Methods The expression levels of Wnt3a and EMT-associated proteins (E-cadherin, vimentin, and β-catenin) were assessed by immunohistochemistry in human colon cancer tissues to evaluate the clinicopathological significance of Wnt3a, as well as the correlation between Wnt3a and EMT. We then upregulated Wnt3a expression in HCT116 colon cancer cells, established a nude mouse xenograft model, detected the expression of EMT and Wnt/β-catenin signaling-associated proteins, and observed invasion and clone-initiating abilities. Results In 203 human colon cancer tissue samples, Wnt3a protein overexpression was related to colon cancer histological differentiation (P = 0.004), clinical stage (P = 0.008), presence of metastasis and recurrence (P = 0.036), and survival time (P = 0.007) of colon cancer patients. Wnt3a expression was notably concomitant with EMT immunohistochemical features, such as reduced expression of the epithelial marker E-cadherin (P = 0.012), increased expression of the mesenchymal marker vimentin (P = 0.002), and cytoplasmic distribution of β-catenin (P = 0.021). Results of in vitro and in vivo experiments showed that Wnt3a overexpression could alter cell morphology, regulate EMT-associated protein expression, and enhance clone-initiation and invasion. Dkk1 (antagonist of Wnt/β-catenin signaling) could also partially reverse the expression of EMT-associated proteins in Wnt3a-overexpressing cells. Conclusions Wnt3a expression was associated with EMT and promoted colon cancer progression. The EMT-inducing effect was partially due to the stimulative effect of Wnt3a on the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,Department of Pathology, Tianjin Medical University, Tianjin, 300070, China. .,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Zhiyong Liu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Runfen Cheng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Yixian Li
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
40
|
Schäfer G, Narasimha M, Vogelsang E, Leptin M. Cadherin switching during the formation and differentiation of the Drosophila mesoderm – implications for epithelial-to-mesenchymal transitions. Development 2014. [DOI: 10.1242/dev.110882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|