1
|
Latham ZD, Bermudez A, Hu JK, Lin NYC. Regulation of epithelial cell jamming transition by cytoskeleton and cell-cell interactions. BIOPHYSICS REVIEWS 2024; 5:041301. [PMID: 39416285 PMCID: PMC11479637 DOI: 10.1063/5.0220088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Multicellular systems, such as epithelial cell collectives, undergo transitions similar to those in inert physical systems like sand piles and foams. To remodel or maintain tissue organization during development or disease, these collectives transition between fluid-like and solid-like states, undergoing jamming or unjamming transitions. While these transitions share principles with physical systems, understanding their regulation and implications in cell biology is challenging. Although cell jamming and unjamming follow physics principles described by the jamming diagram, they are fundamentally biological processes. In this review, we explore how cellular processes and interactions regulate jamming and unjamming transitions. We begin with an overview of how these transitions control tissue remodeling in epithelial model systems and describe recent findings of the physical principles governing tissue solidification and fluidization. We then explore the mechanistic pathways that modulate the jamming phase diagram axes, focusing on the regulation of cell fluctuations and geometric compatibility. Drawing upon seminal works in cell biology, we discuss the roles of cytoskeleton and cell-cell adhesion in controlling cell motility and geometry. This comprehensive view illustrates the molecular control of cell jamming and unjamming, crucial for tissue remodeling in various biological contexts.
Collapse
Affiliation(s)
- Zoe D. Latham
- Bioengineering Department, UCLA, Los Angeles, California 90095, USA
| | | | - Jimmy K. Hu
- Authors to whom correspondence should be addressed: and
| | | |
Collapse
|
2
|
Hu B, Pinzour J, Patel A, Rooney F, Zerwic A, Gao Y, Nguyen NT, Xie H, Ye D, Lin F. Gα13 controls pharyngeal endoderm convergence by regulating E-cadherin expression and RhoA activation. Development 2024; 151:dev202597. [PMID: 39258889 PMCID: PMC11463957 DOI: 10.1242/dev.202597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 08/28/2024] [Indexed: 09/12/2024]
Abstract
Pharyngeal endoderm cells undergo convergence and extension (C&E), which is essential for endoderm pouch formation and craniofacial development. Our previous work implicates Gα13/RhoA-mediated signaling in regulating this process, but the underlying mechanisms remain unclear. Here, we have used endoderm-specific transgenic and Gα13 mutant zebrafish to demonstrate that Gα13 plays a crucial role in pharyngeal endoderm C&E by regulating RhoA activation and E-cadherin expression. We showed that during C&E, endodermal cells gradually establish stable cell-cell contacts, acquire apical-basal polarity and undergo actomyosin-driven apical constriction, which are processes that require Gα13. Additionally, we found that Gα13-deficient embryos exhibit reduced E-cadherin expression, partially contributing to endoderm C&E defects. Notably, interfering with RhoA function disrupts spatial actomyosin activation without affecting E-cadherin expression. Collectively, our findings identify crucial cellular processes for pharyngeal endoderm C&E and reveal that Gα13 controls this through two independent pathways - modulating RhoA activation and regulating E-cadherin expression - thus unveiling intricate mechanisms governing pharyngeal endoderm morphogenesis.
Collapse
Affiliation(s)
- Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Joshua Pinzour
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Asmi Patel
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Faith Rooney
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amie Zerwic
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nhan T. Nguyen
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Huaping Xie
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ding Ye
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
3
|
Mozin E, Massouridès E, Mournetas V, Lièvre C, Bourdon A, Jackson DL, Packer JS, Seong J, Trapnell C, Le Guiner C, Adjali O, Pinset C, Mack DL, Dupont JB. Dystrophin deficiency impairs cell junction formation during embryonic myogenesis from pluripotent stem cells. iScience 2024; 27:110242. [PMID: 39040067 PMCID: PMC11261405 DOI: 10.1016/j.isci.2024.110242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/02/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Mutations in the DMD gene lead to Duchenne muscular dystrophy (DMD), a severe neuromuscular disorder affecting young boys as they acquire motor functions. DMD is typically diagnosed at 2-4 years of age, but the absence of dystrophin has negative impacts on skeletal muscles before overt symptoms appear in patients, which poses a serious challenge in current standards of care. Here, we investigated the consequences of dystrophin deficiency during skeletal muscle development. We used single-cell transcriptome profiling to characterize the myogenic trajectory of human pluripotent stem cells and showed that DMD cells bifurcate to an alternative branch when they reach the somite stage. Dystrophin deficiency was linked to marked dysregulations of cell junction proteins involved in the cell state transitions characteristic of embryonic somitogenesis. Altogether, this work demonstrates that in vitro, dystrophin deficiency has deleterious effects on cell-cell communication during myogenic development, which should be considered in future therapeutic strategies for DMD.
Collapse
Affiliation(s)
- Elise Mozin
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | | | | | - Clémence Lièvre
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Audrey Bourdon
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Dana L. Jackson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Jonathan S. Packer
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Juyoung Seong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98105, USA
| | | | - Oumeya Adjali
- Nantes Université, CHU Nantes, INSERM, TARGET, F-44000 Nantes, France
| | - Christian Pinset
- Centre d’Etude des Cellules Souches, I-Stem, AFM, F-91100 Corbeil-Essonnes, France
| | - David L. Mack
- Institute for Stem Cell and Regenerative Medicine, Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | | |
Collapse
|
4
|
Chouhan G, Lewis NS, Ghanekar V, Koti Ainavarapu SR, Inamdar MM, Sonawane M. Cell-size-dependent regulation of Ezrin dictates epithelial resilience to stretch by countering myosin-II-mediated contractility. Cell Rep 2024; 43:114271. [PMID: 38823013 DOI: 10.1016/j.celrep.2024.114271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 04/22/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024] Open
Abstract
The epithelial adaptations to mechanical stress are facilitated by molecular and tissue-scale changes that include the strengthening of junctions, cytoskeletal reorganization, and cell-proliferation-mediated changes in tissue rheology. However, the role of cell size in controlling these properties remains underexplored. Our experiments in the zebrafish embryonic epidermis, guided by theoretical estimations, reveal a link between epithelial mechanics and cell size, demonstrating that an increase in cell size compromises the tissue fracture strength and compliance. We show that an increase in E-cadherin levels in the proliferation-deficient epidermis restores epidermal compliance but not the fracture strength, which is largely regulated by Ezrin-an apical membrane-cytoskeleton crosslinker. We show that Ezrin fortifies the epithelium in a cell-size-dependent manner by countering non-muscle myosin-II-mediated contractility. This work uncovers the importance of cell size maintenance in regulating the mechanical properties of the epithelium and fostering protection against future mechanical stresses.
Collapse
Affiliation(s)
- Geetika Chouhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Natasha Steffi Lewis
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Vallari Ghanekar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | | | - Mandar M Inamdar
- Department of Civil Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India.
| |
Collapse
|
5
|
Arslan FN, Hannezo É, Merrin J, Loose M, Heisenberg CP. Adhesion-induced cortical flows pattern E-cadherin-mediated cell contacts. Curr Biol 2024; 34:171-182.e8. [PMID: 38134934 DOI: 10.1016/j.cub.2023.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/25/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Metazoan development relies on the formation and remodeling of cell-cell contacts. Dynamic reorganization of adhesion receptors and the actomyosin cell cortex in space and time plays a central role in cell-cell contact formation and maturation. Nevertheless, how this process is mechanistically achieved when new contacts are formed remains unclear. Here, by building a biomimetic assay composed of progenitor cells adhering to supported lipid bilayers functionalized with E-cadherin ectodomains, we show that cortical F-actin flows, driven by the depletion of myosin-2 at the cell contact center, mediate the dynamic reorganization of adhesion receptors and cell cortex at the contact. E-cadherin-dependent downregulation of the small GTPase RhoA at the forming contact leads to both a depletion of myosin-2 and a decrease of F-actin at the contact center. At the contact rim, in contrast, myosin-2 becomes enriched by the retraction of bleb-like protrusions, resulting in a cortical tension gradient from the contact rim to its center. This tension gradient, in turn, triggers centrifugal F-actin flows, leading to further accumulation of F-actin at the contact rim and the progressive redistribution of E-cadherin from the contact center to the rim. Eventually, this combination of actomyosin downregulation and flows at the contact determines the characteristic molecular organization, with E-cadherin and F-actin accumulating at the contact rim, where they are needed to mechanically link the contractile cortices of the adhering cells.
Collapse
Affiliation(s)
- Feyza Nur Arslan
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria; Institute of Bioengineering, École polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Édouard Hannezo
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Jack Merrin
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Martin Loose
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | | |
Collapse
|
6
|
Concha ML, Reig G. Origin, form and function of extraembryonic structures in teleost fishes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210264. [PMID: 36252221 PMCID: PMC9574637 DOI: 10.1098/rstb.2021.0264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022] Open
Abstract
Teleost eggs have evolved a highly derived early developmental pattern within vertebrates as a result of the meroblastic cleavage pattern, giving rise to a polar stratified architecture containing a large acellular yolk and a small cellular blastoderm on top. Besides the acellular yolk, the teleost-specific yolk syncytial layer (YSL) and the superficial epithelial enveloping layer are recognized as extraembryonic structures that play critical roles throughout embryonic development. They provide enriched microenvironments in which molecular feedback loops, cellular interactions and mechanical signals emerge to sculpt, among other things, embryonic patterning along the dorsoventral and left-right axes, mesendodermal specification and the execution of morphogenetic movements in the early embryo and during organogenesis. An emerging concept points to a critical role of extraembryonic structures in reinforcing early genetic and morphogenetic programmes in reciprocal coordination with the embryonic blastoderm, providing the necessary boundary conditions for development to proceed. In addition, the role of the enveloping cell layer in providing mechanical, osmotic and immunological protection during early stages of development, and the autonomous nutritional support provided by the yolk and YSL, have probably been key aspects that have enabled the massive radiation of teleosts to colonize every ecological niche on the Earth. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Miguel L. Concha
- Integrative Biology Program, Institute of Biomedical Sciences (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Biomedical Neuroscience Institute (BNI), Santiago 8380453, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
| | - Germán Reig
- Escuela de Tecnología Médica y del Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 7800003, Chile
| |
Collapse
|
7
|
Shan B, Horton EC, Xu SC, Huntington KE, Kawano DK, Mendoza CL, Lin L, Stafford CM, Allen ED, Huang J, Nakahara H, Greenstein LE, Hille MB. Dephosphorylation of Y228 and Y217 and phosphorylation of Y335 in p120 catenin activate convergent extension during zebrafish gastrulation. Dev Dyn 2022; 251:1934-1951. [PMID: 35996230 DOI: 10.1002/dvdy.524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/15/2022] [Accepted: 05/26/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The cadherin-associated protein p120 catenin regulates convergent extension through interactions with cadherin proteins, Cdc42, and Rac1, as we previously showed in zebrafish (Danio rerio). Phosphorylation of p120 catenin changes the nature of its activity in vitro but is virtually unexplored in embryos. We used our previously developed antisense RNA splice-site morpholino targeted to endogenous p120 catenin-δ1 to cause defects in axis elongation probing the functions of three p120 catenin tyrosine-phosphorylation sites in gastrulating zebrafish embryos. RESULTS The morpholino-induced defects were rescued by co-injections with mouse p120 catenin-δ1-3A mRNAs mutated at residues Y228 and Y217 to a non-phosphorylatable phenylalanine (F) or mutated at residue Y335 to a phosphomimetic glutamic acid (E). Co-injection of the complementary mutations Y228E, Y217E, or Y335F mRNAs partially rescued embryos whereas dual mutation to Y228E-Y217E blocked rescue. Immunopurification showed Y228F mutant proteins preferentially interacted with Rac1, potentially promoting cell migration. In contrast, the phosphomimetic Y228E preferentially interacted with E-cadherin increasing adhesion. Both Y228F and Y335F strongly bind VAV2. CONCLUSIONS p120 catenin serves dual roles during gastrulation of zebrafish. Phosphorylation and dephosphorylation of tyrosine residues Y217, Y228, and Y335 precisely balance cell adhesion and cell migration to facilitate somite compaction and axis elongation.
Collapse
Affiliation(s)
- Botao Shan
- Department of Biology, University of Washington, Seattle, Washington, USA.,Tulane University School of Medicine, New Orleans, LA, USA
| | - Emma C Horton
- Department of Biology, University of Washington, Seattle, Washington, USA.,Developmental and Stem Cell Biology Program, University of California San Francisco, San Francisco, CA, USA
| | - Shan C Xu
- Department of Biology, University of Washington, Seattle, Washington, USA.,New York University Stern Business School, New York, NY, USA
| | - Kelsey E Huntington
- Department of Biology, University of Washington, Seattle, Washington, USA.,Pathobiology Graduate Program, Division of Biology and Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Dane K Kawano
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Biology, Stanford University, Stanford, CA, USA
| | - Clemence L Mendoza
- Department of Biology, University of Washington, Seattle, Washington, USA.,VA Portland Health Care System, Portland, OR, USA
| | - Laura Lin
- Department of Biology, University of Washington, Seattle, Washington, USA.,Touro University California College of Osteopathic Medicine, Vallejo, CA, USA
| | | | - Emili D Allen
- Department of Biology, University of Washington, Seattle, Washington, USA.,Adaptive Biotechnologies Corp, Seattle, WA, USA
| | - Joyce Huang
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - Hiroko Nakahara
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Lewis E Greenstein
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Medical Entomology, Champaign, IL, USA
| | - Merrill B Hille
- Department of Biology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
8
|
Kupai A, Nakahara H, Voss KM, Hirano MS, Rodriguez A, Lackey DL, Murayama JF, Mathieson CJ, Shan B, Horton EC, Curtis GH, Huang J, Hille MB. Phosphorylation of serine residues S252, S268/S269, and S879 in p120 catenin activates migration of presomitic mesoderm in gastrulating zebrafish embryos. Dev Dyn 2022; 251:1952-1967. [PMID: 35706088 DOI: 10.1002/dvdy.508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/15/2022] [Accepted: 05/26/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Cadherin-associated protein p120 catenin regulates cell adhesion and migration in cell cultures and is required for axial elongation in embryos. Its roles in adhesion and cell migration are regulated by phosphorylation. We determined the effects of phosphorylation of six serine and three threonine residues in p120 catenin during zebrafish (Danio rerio) embryogenesis. RESULTS We knocked down endogenous p120 catenin-δ1 with an antisense RNA-splice-site morpholino (Sp-MO) causing defects in axis elongation. These defects were rescued by co-injections of mRNAs for wildtype mouse p120 catenin-δ1-3A or various mutated forms. Several mRNAs containing serine or threonine codons singly or doubly mutated to phosphomimetic glutamic acid rescued, and some nonphosphorylatable mutants did not. CONCLUSIONS We discovered that phosphorylation of serine residue S252 or S879 is required for convergent extension of zebrafish embryos, since rescue occurred only when these residues were mutated to glutamic acid. In addition, the phosphorylation of either S268 or S269 is required, not both, consistent with the presence of only a single one of these residues in two isoforms of zebrafish and Xenopus laevis. In summary, phosphorylation of multiple serine and threonine residues of p120 catenin activates migration of presomitic mesoderm of zebrafish embryos facilitating elongation of the dorsal axis.
Collapse
Affiliation(s)
- Ariana Kupai
- Department of Biology, University of Washington, Seattle, Washington, USA.,Van Andel Institute, Grand Rapids, Michigan, USA
| | - Hiroko Nakahara
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Kathleen M Voss
- Department of Biology, University of Washington, Seattle, Washington, USA.,Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew S Hirano
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Electrical and Computer Engineering, University of Washington, Seattle, Washington, USA
| | - Alexis Rodriguez
- Department of Biology, University of Washington, Seattle, Washington, USA.,Apex Systems, Santa Clara, California, USA
| | - Donna L Lackey
- Department of Biology, University of Washington, Seattle, Washington, USA.,PACT Pharma, 2 Corporate Drive, South San Francisco, California, USA
| | - James F Murayama
- Department of Biology, University of Washington, Seattle, Washington, USA.,DDS Private Practice, Mission Viejo, California, USA
| | - Chase J Mathieson
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Integrative Biomedical and Diagnostic Science, Oregon Health and Science University, Portland, Oregon, USA
| | - Botao Shan
- Department of Biology, University of Washington, Seattle, Washington, USA.,Tulane University Medical School, New Orleans, Louisiana, USA
| | - Emma C Horton
- Department of Biology, University of Washington, Seattle, Washington, USA.,Developmental & Stem Cell Biology Program, University of California San Francisco, San Francisco, California, USA
| | - Grace H Curtis
- Department of Biology, University of Washington, Seattle, Washington, USA.,School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Joyce Huang
- Department of Biology, University of Washington, Seattle, Washington, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, California, USA
| | - Merrill B Hille
- Department of Biology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Manikandan P, Sarmah S, Marrs JA. Ethanol Effects on Early Developmental Stages Studied Using the Zebrafish. Biomedicines 2022; 10:2555. [PMID: 36289818 PMCID: PMC9599251 DOI: 10.3390/biomedicines10102555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) results from prenatal ethanol exposure. The zebrafish (Danio rerio) is an outstanding in vivo FASD model. Early development produced the three germ layers and embryonic axes patterning. A critical pluripotency transcriptional gene circuit of sox2, pou5f1 (oct4; recently renamed pou5f3), and nanog maintain potency and self-renewal. Ethanol affects sox2 expression, which functions with pou5f1 to control target gene transcription. Various genes, like elf3, may interact and regulate sox2, and elf3 knockdown affects early development. Downstream of the pluripotency transcriptional circuit, developmental signaling activities regulate morphogenetic cell movements and lineage specification. These activities are also affected by ethanol exposure. Hedgehog signaling is a critical developmental signaling pathway that controls numerous developmental events, including neural axis specification. Sonic hedgehog activities are affected by embryonic ethanol exposure. Activation of sonic hedgehog expression is controlled by TGF-ß family members, Nodal and Bmp, during dorsoventral (DV) embryonic axis establishment. Ethanol may perturb TGF-ß family receptors and signaling activities, including the sonic hedgehog pathway. Significantly, experiments show that activation of sonic hedgehog signaling rescues some embryonic ethanol exposure effects. More research is needed to understand how ethanol affects early developmental signaling and morphogenesis.
Collapse
Affiliation(s)
| | | | - James A. Marrs
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Guidance by followers ensures long-range coordination of cell migration through α-catenin mechanoperception. Dev Cell 2022; 57:1529-1544.e5. [PMID: 35613615 DOI: 10.1016/j.devcel.2022.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 03/09/2022] [Accepted: 05/02/2022] [Indexed: 11/23/2022]
Abstract
Morphogenesis, wound healing, and some cancer metastases depend upon the migration of cell collectives that need to be guided to their destination as well as coordinated with other cell movements. During zebrafish gastrulation, the extension of the embryonic axis is led by the mesendodermal polster that migrates toward the animal pole, followed by the axial mesoderm that undergoes convergence and extension. Here, we investigate how polster cells are guided toward the animal pole. Using a combination of precise laser ablations, advanced transplants, and functional as well as in silico approaches, we establish that each polster cell is oriented by its immediate follower cells. Each cell perceives the migration of followers, through E-cadherin/α-catenin mechanotransduction, and aligns with them. Therefore, directional information propagates from cell to cell over the whole tissue. Such guidance of migrating cells by followers ensures long-range coordination of movements and developmental robustness.
Collapse
|
11
|
Chen PH, Tjong WY, Yang HC, Liu HY, Stern A, Chiu DTY. Glucose-6-Phosphate Dehydrogenase, Redox Homeostasis and Embryogenesis. Int J Mol Sci 2022; 23:ijms23042017. [PMID: 35216131 PMCID: PMC8878822 DOI: 10.3390/ijms23042017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Normal embryogenesis requires complex regulation and precision, which depends on multiple mechanistic details. Defective embryogenesis can occur by various mechanisms. Maintaining redox homeostasis is of importance during embryogenesis. NADPH, as produced from the action of glucose-6-phosphate dehydrogenase (G6PD), has an important role in redox homeostasis, serving as a cofactor for glutathione reductase in the recycling of glutathione from oxidized glutathione and for NADPH oxidases and nitric oxide synthases in the generation of reactive oxygen (ROS) and nitrogen species (RNS). Oxidative stress differentially influences cell fate and embryogenesis. While low levels of stress (eustress) by ROS and RNS promote cell growth and differentiation, supra-physiological concentrations of ROS and RNS can lead to cell demise and embryonic lethality. G6PD-deficient cells and organisms have been used as models in embryogenesis for determining the role of redox signaling in regulating cell proliferation, differentiation and migration. Embryogenesis is also modulated by anti-oxidant enzymes, transcription factors, microRNAs, growth factors and signaling pathways, which are dependent on redox regulation. Crosstalk among transcription factors, microRNAs and redox signaling is essential for embryogenesis.
Collapse
Affiliation(s)
- Po-Hsiang Chen
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Wen-Ye Tjong
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Correspondence: ; Tel.: +886-3-6108175; Fax: +886-3-6102327
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, NY 10016, USA;
| | - Daniel Tsun-Yee Chiu
- Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; (P.-H.C.); (W.-Y.T.); (D.T.-Y.C.)
| |
Collapse
|
12
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
13
|
Mishra N, Heisenberg CP. Dissecting Organismal Morphogenesis by Bridging Genetics and Biophysics. Annu Rev Genet 2021; 55:209-233. [PMID: 34460295 DOI: 10.1146/annurev-genet-071819-103748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multicellular organisms develop complex shapes from much simpler, single-celled zygotes through a process commonly called morphogenesis. Morphogenesis involves an interplay between several factors, ranging from the gene regulatory networks determining cell fate and differentiation to the mechanical processes underlying cell and tissue shape changes. Thus, the study of morphogenesis has historically been based on multidisciplinary approaches at the interface of biology with physics and mathematics. Recent technological advances have further improved our ability to study morphogenesis by bridging the gap between the genetic and biophysical factors through the development of new tools for visualizing, analyzing, and perturbing these factors and their biochemical intermediaries. Here, we review how a combination of genetic, microscopic, biophysical, and biochemical approaches has aided our attempts to understand morphogenesis and discuss potential approaches that may be beneficial to such an inquiry in the future. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Nikhil Mishra
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria; ,
| | | |
Collapse
|
14
|
Petridou NI, Corominas-Murtra B, Heisenberg CP, Hannezo E. Rigidity percolation uncovers a structural basis for embryonic tissue phase transitions. Cell 2021; 184:1914-1928.e19. [PMID: 33730596 PMCID: PMC8055543 DOI: 10.1016/j.cell.2021.02.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/09/2020] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
Embryo morphogenesis is impacted by dynamic changes in tissue material properties, which have been proposed to occur via processes akin to phase transitions (PTs). Here, we show that rigidity percolation provides a simple and robust theoretical framework to predict material/structural PTs of embryonic tissues from local cell connectivity. By using percolation theory, combined with directly monitoring dynamic changes in tissue rheology and cell contact mechanics, we demonstrate that the zebrafish blastoderm undergoes a genuine rigidity PT, brought about by a small reduction in adhesion-dependent cell connectivity below a critical value. We quantitatively predict and experimentally verify hallmarks of PTs, including power-law exponents and associated discontinuities of macroscopic observables. Finally, we show that this uniform PT depends on blastoderm cells undergoing meta-synchronous divisions causing random and, consequently, uniform changes in cell connectivity. Collectively, our theoretical and experimental findings reveal the structural basis of material PTs in an organismal context.
Collapse
Affiliation(s)
| | | | | | - Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
15
|
Grimaldi C, Schumacher I, Boquet-Pujadas A, Tarbashevich K, Vos BE, Bandemer J, Schick J, Aalto A, Olivo-Marin JC, Betz T, Raz E. E-cadherin focuses protrusion formation at the front of migrating cells by impeding actin flow. Nat Commun 2020; 11:5397. [PMID: 33106478 PMCID: PMC7588466 DOI: 10.1038/s41467-020-19114-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 09/25/2020] [Indexed: 12/11/2022] Open
Abstract
The migration of many cell types relies on the formation of actomyosin-dependent protrusions called blebs, but the mechanisms responsible for focusing this kind of protrusive activity to the cell front are largely unknown. Here, we employ zebrafish primordial germ cells (PGCs) as a model to study the role of cell-cell adhesion in bleb-driven single-cell migration in vivo. Utilizing a range of genetic, reverse genetic and mathematical tools, we define a previously unknown role for E-cadherin in confining bleb-type protrusions to the leading edge of the cell. We show that E-cadherin-mediated frictional forces impede the backwards flow of actomyosin-rich structures that define the domain where protrusions are preferentially generated. In this way, E-cadherin confines the bleb-forming region to a restricted area at the cell front and reinforces the front-rear axis of migrating cells. Accordingly, when E-cadherin activity is reduced, the bleb-forming area expands, thus compromising the directional persistence of the cells. The arrival of migratory cells at their targets relies on following precise routes within tissues. Here the authors demonstrate that the cell adhesion molecule E-cadherin can control the path of cell migration by confining the site where bleb-type protrusions form within the cell front.
Collapse
Affiliation(s)
- Cecilia Grimaldi
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Isabel Schumacher
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Aleix Boquet-Pujadas
- Institut Pasteur, Bioimage Analysis Unit, 75105, Paris, France.,CNRS UMR 3691, 75105, Paris, France.,Sorbonne Université, 75005, Paris, France
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Bart Eduard Vos
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Jan Bandemer
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Jan Schick
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | - Anne Aalto
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany
| | | | - Timo Betz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany.,Institute of Physics - Biophysics, Georg August Universität, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, 48149, Münster, Germany.
| |
Collapse
|
16
|
Banote RK, Chebli J, Şatır TM, Varshney GK, Camacho R, Ledin J, Burgess SM, Abramsson A, Zetterberg H. Amyloid precursor protein-b facilitates cell adhesion during early development in zebrafish. Sci Rep 2020; 10:10127. [PMID: 32576936 PMCID: PMC7311384 DOI: 10.1038/s41598-020-66584-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/21/2020] [Indexed: 01/05/2023] Open
Abstract
Understanding the biological function of amyloid beta (Aβ) precursor protein (APP) beyond its role in Alzheimer's disease is emerging. Yet, its function during embryonic development is poorly understood. The zebrafish APP orthologue, Appb, is strongly expressed during early development but thus far has only been studied via morpholino-mediated knockdown. Zebrafish enables analysis of cellular processes in an ontogenic context, which is limited in many other vertebrates. We characterized zebrafish carrying a homozygous mutation that introduces a premature stop in exon 2 of the appb gene. We report that appb mutants are significantly smaller until 2 dpf and display perturbed enveloping layer (EVL) integrity and cell protrusions at the blastula stage. Moreover, appb mutants surviving beyond 48 hpf exhibited no behavioral defects at 6 dpf and developed into healthy and fertile adults. The expression of the app family member, appa, was also found to be altered in appb mutants. Taken together, we show that appb is involved in the initial development of zebrafish by supporting the integrity of the EVL, likely by mediating cell adhesion properties. The loss of Appb might then be compensated for by other app family members to maintain normal development.
Collapse
Affiliation(s)
- Rakesh Kumar Banote
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, S-41345, Gothenburg, Sweden.,Cellectricon AB, Neongatan 4B, SE-431 53, Mölndal, Sweden
| | - Jasmine Chebli
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, S-41345, Gothenburg, Sweden
| | - Tuğçe Munise Şatır
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, S-41345, Gothenburg, Sweden
| | - Gaurav K Varshney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA.,Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Rafael Camacho
- Centre for Cellular Imaging, Core Facilities, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Ledin
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA.,Department of Organismal Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Alexandra Abramsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, S-41345, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, S-41345, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N3BG, United Kingdom.,UK Dementia Research Institute, London, WC1N3BG, United Kingdom
| |
Collapse
|
17
|
Hatching gland development and hatching in zebrafish embryos: A role for zinc and its transporters Zip10 and Znt1a. Biochem Biophys Res Commun 2020; 528:698-705. [PMID: 32517868 DOI: 10.1016/j.bbrc.2020.05.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 11/22/2022]
Abstract
Zinc transporters of the ZIP (Slc39, importers) and ZnT (Slc30, exporters) protein families have evolutionary conserved roles in biology. The aim of the present study was to explore the role of zinc, and zinc transporters Zip10 and Znt1a in zebrafish hatching gland development and larval hatching. In the study, knockdown of genes for Zip10 and Znt1a in zebrafish embryos was achieved using morpholino-modified oligonucleotides. A partial loss-of-function Znt1a mutant (Znt1asa17) allowed comparison with the Znt1a morphant. Free Zn2+ in embryos and apoptosis were investigated using fluorescent dyes whereas gene expression was investigated by whole-mount in situ hybridization (WISH). The results showed high levels of free Zn2+ in the hatching gland cells (HGC) along with abundant expression of zip10 and znt1a in normal embryo. Knockdown of zip10 reduced free Zn2+ in HGC, ceased their normal developmental apoptosis, and resulted in displacement and later disappearance of hatching glands and hatching enzymes he1a and catL1b, and inability to hatch. Conversely, knockdown of znt1a or the Znt1asa17 mutation accelerated hatching and coincided with high expression of hatching enzymes and free Zn2+ in the HGC. Thus, Zip10 and free Zn2+ in the HGC are required both for their development and function. This study also demonstrated the opposite functions of the two zinc transporters, ZIP10 and ZnT1 as well as shedding light on the role of Zn2+ in regulation of the human hatching enzyme homologue, ovastacin, which is activated by zinc and cleaves the zona pellucida protein, ZP2, to prevent polyspermy.
Collapse
|
18
|
Sarmah S, Srivastava R, McClintick JN, Janga SC, Edenberg HJ, Marrs JA. Embryonic ethanol exposure alters expression of sox2 and other early transcripts in zebrafish, producing gastrulation defects. Sci Rep 2020; 10:3951. [PMID: 32127575 PMCID: PMC7054311 DOI: 10.1038/s41598-020-59043-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023] Open
Abstract
Ethanol exposure during prenatal development causes fetal alcohol spectrum disorder (FASD), the most frequent preventable birth defect and neurodevelopmental disability syndrome. The molecular targets of ethanol toxicity during development are poorly understood. Developmental stages surrounding gastrulation are very sensitive to ethanol exposure. To understand the effects of ethanol on early transcripts during embryogenesis, we treated zebrafish embryos with ethanol during pre-gastrulation period and examined the transcripts by Affymetrix GeneChip microarray before gastrulation. We identified 521 significantly dysregulated genes, including 61 transcription factors in ethanol-exposed embryos. Sox2, the key regulator of pluripotency and early development was significantly reduced. Functional annotation analysis showed enrichment in transcription regulation, embryonic axes patterning, and signaling pathways, including Wnt, Notch and retinoic acid. We identified all potential genomic targets of 25 dysregulated transcription factors and compared their interactions with the ethanol-dysregulated genes. This analysis predicted that Sox2 targeted a large number of ethanol-dysregulated genes. A gene regulatory network analysis showed that many of the dysregulated genes are targeted by multiple transcription factors. Injection of sox2 mRNA partially rescued ethanol-induced gene expression, epiboly and gastrulation defects. Additional studies of this ethanol dysregulated network may identify therapeutic targets that coordinately regulate early development.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Rajneesh Srivastava
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jeanette N McClintick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sarath C Janga
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| |
Collapse
|
19
|
|
20
|
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: 8] [Impact Index Per Article: 1.6] [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
|
21
|
Abstract
Epiboly is a conserved gastrulation movement describing the thinning and spreading of a sheet or multi-layer of cells. The zebrafish embryo has emerged as a vital model system to address the cellular and molecular mechanisms that drive epiboly. In the zebrafish embryo, the blastoderm, consisting of a simple squamous epithelium (the enveloping layer) and an underlying mass of deep cells, as well as a yolk nuclear syncytium (the yolk syncytial layer) undergo epiboly to internalize the yolk cell during gastrulation. The major events during zebrafish epiboly are: expansion of the enveloping layer and the internal yolk syncytial layer, reduction and removal of the yolk membrane ahead of the advancing blastoderm margin and deep cell rearrangements between the enveloping layer and yolk syncytial layer to thin the blastoderm. Here, work addressing the cellular and molecular mechanisms as well as the sources of the mechanical forces that underlie these events is reviewed. The contribution of recent findings to the current model of epiboly as well as open questions and future prospects are also discussed.
Collapse
|
22
|
Rutherford NE, Wong AH, Bruce AEE. Spatiotemporal characterization of dynamic epithelial filopodia during zebrafish epiboly. Dev Dyn 2019; 248:997-1008. [PMID: 31390119 DOI: 10.1002/dvdy.94] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/08/2019] [Accepted: 06/28/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND During zebrafish epiboly, the embryonic cell mass, or blastoderm, spreads to enclose the yolk cell. The blastoderm consists of an outer epithelial sheet, the enveloping layer (EVL), and the underlying deep cell layer (DEL). Studies have provided insights into the mechanisms of EVL and deep cell epiboly, but little is known about the interactions between the two cell layers and what role they may play during epiboly. RESULTS We used live imaging to examine EVL basal protrusions. We identified them as filopodia based on f-actin content and localization of fluorescently tagged filopodial markers. A spatiotemporal analysis revealed that the largest number of EVL filopodia were present during early epiboly at the animal pole. In functional studies, expression of a constitutively active actin-bundling protein resulted in increased filopodial length and delayed gastrulation. CONCLUSIONS We identified protrusions on the basal surface of EVL cells as filopodia and showed that they are present throughout the EVL during epiboly. The largest number of filopodia was at the animal pole during early epiboly, which is when and where deep cell radial intercalations occur to the greatest extent. These findings suggest that EVL filopodia may function during epiboly to promote deep cell rearrangements during epiboly initiation.
Collapse
Affiliation(s)
- Nathan E Rutherford
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario
| | - Alexander H Wong
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario
| | - Ashley E E Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario
| |
Collapse
|
23
|
Cheng XN, Shao M, Shi DL. Collagen triple helix repeat containing 1a (Cthrc1a) regulates cell adhesion and migration during gastrulation in zebrafish. Exp Cell Res 2019; 381:112-120. [DOI: 10.1016/j.yexcr.2019.04.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/29/2019] [Indexed: 01/27/2023]
|
24
|
Sampedro MF, Izaguirre MF, Sigot V. E-cadherin expression pattern during zebrafish embryonic epidermis development. F1000Res 2019; 7:1489. [PMID: 30473778 PMCID: PMC6234749 DOI: 10.12688/f1000research.15932.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2019] [Indexed: 12/20/2022] Open
Abstract
Background: E-cadherin is the major adhesion receptor in epithelial adherens junctions (AJs). On established epidermis, E-cadherin performs fine-tuned cell-cell contact remodeling to maintain tissue integrity, which is characterized by modulation of cell shape, size and packing density. In zebrafish, the organization and distribution of E-cadherin in AJs during embryonic epidermis development remain scarcely described. Methods: Combining classical immunofluorescence, deconvolution microscopy and 3D-segmentation of AJs in epithelial cells, a quantitative approach was implemented to assess the spatial and temporal distribution of E-cadherin across zebrafish epidermis between 24 and 72 hpf. Results: increasing levels of E-cadh protein parallel higher cell density and the appearance of hexagonal cells in the enveloping layer (EVL) as well as the establishments of new cell-cell contacts in the epidermal basal layer (EBL), being significantly between 31 and 48 hpf
. Conclusions: Increasing levels of E-cadherin in AJs correlates with extensive changes in cell morphology towards hexagonal packing during the epidermis morphogenesis.
Collapse
Affiliation(s)
- María Florencia Sampedro
- Laboratorio de Microscopía Aplicada a Estudios Moleculares y Celulares (LAMAE), Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, 3100, Argentina.,Instituto de Investigación y Desarrollo en Bioingeniería y Bioinformática (IBB-CONICET- Universidad Nacional de Entre Ríos), Oro Verde, 3100, Argentina
| | - María Fernanda Izaguirre
- Laboratorio de Microscopía Aplicada a Estudios Moleculares y Celulares (LAMAE), Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, 3100, Argentina
| | - Valeria Sigot
- Laboratorio de Microscopía Aplicada a Estudios Moleculares y Celulares (LAMAE), Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde, 3100, Argentina.,Instituto de Investigación y Desarrollo en Bioingeniería y Bioinformática (IBB-CONICET- Universidad Nacional de Entre Ríos), Oro Verde, 3100, Argentina
| |
Collapse
|
25
|
Mendieta-Serrano MA, Mendez-Cruz FJ, Antúnez-Mojica M, Schnabel D, Alvarez L, Cárdenas L, Lomelí H, Ruiz-Santiesteban JA, Salas-Vidal E. NADPH-Oxidase-derived reactive oxygen species are required for cytoskeletal organization, proper localization of E-cadherin and cell motility during zebrafish epiboly. Free Radic Biol Med 2019; 130:82-98. [PMID: 30342187 DOI: 10.1016/j.freeradbiomed.2018.10.416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022]
Abstract
Cell movements are essential for morphogenesis during animal development. Epiboly is the first morphogenetic process in zebrafish in which cells move en masse to thin and spread the deep and enveloping cell layers of the blastoderm over the yolk cell. While epiboly has been shown to be controlled by complex molecular networks, the contribution of reactive oxygen species (ROS) to this process has not previously been studied. Here, we show that ROS are required for epiboly in zebrafish. Visualization of ROS in whole embryos revealed dynamic patterns during epiboly progression. Significantly, inhibition of NADPH oxidase activity leads to a decrease in ROS formation, delays epiboly, alters E-cadherin and cytoskeleton patterns and, by 24 h post-fertilization, decreases embryo survival, effects that are rescued by hydrogen peroxide treatment. Our findings suggest that a delicate ROS balance is required during early development and that disruption of that balance interferes with cell adhesion, leading to defective cell motility and epiboly progression.
Collapse
Affiliation(s)
| | | | - Mayra Antúnez-Mojica
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62209, Mexico
| | - Denhi Schnabel
- Departamento de Genética del Desarrollo y Fisiología Molecular, Mexico
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62209, Mexico
| | - Luis Cárdenas
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad #2001, Colonia Chamilpa, Cuernavaca, Morelos C.P. 62210, Mexico
| | - Hilda Lomelí
- Departamento de Genética del Desarrollo y Fisiología Molecular, Mexico
| | | | | |
Collapse
|
26
|
Dasgupta S, Cheng V, Vliet SMF, Mitchell CA, Volz DC. Tris(1,3-dichloro-2-propyl) Phosphate Exposure During the Early-Blastula Stage Alters the Normal Trajectory of Zebrafish Embryogenesis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:10820-10828. [PMID: 30157643 PMCID: PMC6169527 DOI: 10.1021/acs.est.8b03730] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) is an organophosphate flame retardant used around the world. Within zebrafish, we previously showed that initiation of TDCIPP exposure during cleavage (0.75 h post-fertilization, hpf) results in epiboly disruption at 6 hpf, leading to dorsalized embryos by 24 hpf, a phenotype that mimics the effects of dorsomorphin (DMP), a bone morphogenetic protein (BMP) antagonist that dorsalizes embryos in the absence of epiboly defects. The objective of this study was to (1) investigate the role of BMP signaling in TDCIPP-induced toxicity during early embryogenesis, (2) identify other pathways and processes targeted by TDCIPP, and (3) characterize the downstream impacts of early developmental defects. Using zebrafish as a model, we first identified a sensitive window for TDCIPP-induced effects following exposure initiation at 0.75 hpf. We then investigated the effects of TDCIPP on the transcriptome during the first 24 h of development using mRNA sequencing and amplicon sequencing. Finally, we relied on whole-mount immunohistochemistry, dye-based labeling, and morphological assessments to study abnormalities later in embryonic development. Overall, our data suggest that the initiation of TDCIPP exposure during early blastula alters the normal trajectory of early embryogenesis by inducing gastrulation defects and aberrant germ-layer formation, leading to abnormal tissue and organ development within the embryo.
Collapse
Affiliation(s)
- Subham Dasgupta
- Department of Environmental Sciences, University of California, Riverside, California 92521, United States
| | - Vanessa Cheng
- Department of Environmental Sciences, University of California, Riverside, California 92521, United States
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
| | - Sara M. F. Vliet
- Department of Environmental Sciences, University of California, Riverside, California 92521, United States
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
| | - Constance A. Mitchell
- Department of Environmental Sciences, University of California, Riverside, California 92521, United States
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
| | - David C. Volz
- Department of Environmental Sciences, University of California, Riverside, California 92521, United States
- Phone: (951) 827-4450; Fax: (951) 827 3993;
| |
Collapse
|
27
|
Wu YH, Lee YH, Shih HY, Chen SH, Cheng YC, Tsun-Yee Chiu D. Glucose-6-phosphate dehydrogenase is indispensable in embryonic development by modulation of epithelial-mesenchymal transition via the NOX/Smad3/miR-200b axis. Cell Death Dis 2018; 9:10. [PMID: 29317613 PMCID: PMC5849038 DOI: 10.1038/s41419-017-0005-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/02/2017] [Accepted: 10/02/2017] [Indexed: 12/24/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is a housekeeping enzyme involved in the pentose phosphate shunt for producing nicotinamide adenine dinucleotide phosphate (NADPH). Severe G6PD deficiency leads to embryonic lethality, but the underlying mechanism is unclear. In the current study, the effects of G6PD on epithelial-mesenchymal transition (EMT), especially during embryonic development, were investigated. The knockdown of G6PD induced morphological changes, accompanied by the suppression of epithelial markers, E-cadherin and β-catenin, in A549 and MDCK cells. Such modulation of EMT was corroborated by the enhancement of migration ability in G6PD-knockdown A549 cells. Zebrafish embryos with g6pd knockdown exhibited downregulation of the E-cadherin/β-catenin adhesion molecules and impaired embryonic development through reduction in epiboly rate and increase in cell shedding at the embryo surface. The dysregulation in zebrafish embryonic development caused by g6pd knockdown could be rescued through human G6PD or CDH1 (E-cadherin gene) cRNA coinjection. The Smad3/miR-200b axis was dysregulated upon G6PD knockdown, and the reconstitution of SMAD3 in G6PD-knockdown A549 cells restored the expression of E-cadherin/β-catenin. The inhibition of NADPH oxidase (NOX) activation through the loss of p22phox signaling was involved in the dysregulation of the Smad3/miR-200b axis upon G6PD knockdown. The reconstitution of G6PD led to the recovery of the regulation of NOX/Smad3/miR-200b signaling and increased the expression of E-cadherin/β-catenin in G6PD-knockdown cells. Thus, these results suggest that in the EMT process, G6PD plays an important regulatory role as an integral component of the NOX/Smad3/miR-200b axis.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Hsuan Lee
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Yu Shih
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsiang Chen
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan.
| | - Daniel Tsun-Yee Chiu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan. .,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
28
|
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: 1.6] [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
|
29
|
Mib1 contributes to persistent directional cell migration by regulating the Ctnnd1-Rac1 pathway. Proc Natl Acad Sci U S A 2017; 114:E9280-E9289. [PMID: 29078376 DOI: 10.1073/pnas.1712560114] [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] [Indexed: 11/18/2022] Open
Abstract
Persistent directional cell migration is involved in animal development and diseases. The small GTPase Rac1 is involved in F-actin and focal adhesion dynamics. Local Rac1 activity is required for persistent directional migration, whereas global, hyperactivated Rac1 enhances random cell migration. Therefore, precise control of Rac1 activity is important for proper directional cell migration. However, the molecular mechanism underlying the regulation of Rac1 activity in persistent directional cell migration is not fully understood. Here, we show that the ubiquitin ligase mind bomb 1 (Mib1) is involved in persistent directional cell migration. We found that knockdown of MIB1 led to an increase in random cell migration in HeLa cells in a wound-closure assay. Furthermore, we explored novel Mib1 substrates for cell migration and found that Mib1 ubiquitinates Ctnnd1. Mib1-mediated ubiquitination of Ctnnd1 K547 attenuated Rac1 activation in cultured cells. In addition, we found that posterior lateral line primordium cells in the zebrafish mib1ta52b mutant showed increased random migration and loss of directional F-actin-based protrusion formation. Knockdown of Ctnnd1 partially rescued posterior lateral line primordium cell migration defects in the mib1ta52b mutant. Taken together, our data suggest that Mib1 plays an important role in cell migration and that persistent directional cell migration is regulated, at least in part, by the Mib1-Ctnnd1-Rac1 pathway.
Collapse
|
30
|
Barone V, Lang M, Krens SFG, Pradhan SJ, Shamipour S, Sako K, Sikora M, Guet CC, Heisenberg CP. An Effective Feedback Loop between Cell-Cell Contact Duration and Morphogen Signaling Determines Cell Fate. Dev Cell 2017; 43:198-211.e12. [PMID: 29033362 DOI: 10.1016/j.devcel.2017.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 11/19/2022]
Abstract
Cell-cell contact formation constitutes an essential step in evolution, leading to the differentiation of specialized cell types. However, remarkably little is known about whether and how the interplay between contact formation and fate specification affects development. Here, we identify a positive feedback loop between cell-cell contact duration, morphogen signaling, and mesendoderm cell-fate specification during zebrafish gastrulation. We show that long-lasting cell-cell contacts enhance the competence of prechordal plate (ppl) progenitor cells to respond to Nodal signaling, required for ppl cell-fate specification. We further show that Nodal signaling promotes ppl cell-cell contact duration, generating a positive feedback loop between ppl cell-cell contact duration and cell-fate specification. Finally, by combining mathematical modeling and experimentation, we show that this feedback determines whether anterior axial mesendoderm cells become ppl or, instead, turn into endoderm. Thus, the interdependent activities of cell-cell signaling and contact formation control fate diversification within the developing embryo.
Collapse
Affiliation(s)
- Vanessa Barone
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Moritz Lang
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria.
| | - S F Gabriel Krens
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Saurabh J Pradhan
- Indian Institute of Science, Education and Research (IISER), Pune 411008, India
| | - Shayan Shamipour
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Keisuke Sako
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Mateusz Sikora
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Călin C Guet
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | | |
Collapse
|
31
|
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: 2.6] [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
|
32
|
Mochizuki T, Luo YJ, Tsai HF, Hagiwara A, Masai I. Cell division and cadherin-mediated adhesion regulate lens epithelial cell movement in zebrafish. Development 2017; 144:708-719. [PMID: 28196805 DOI: 10.1242/dev.138909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/04/2017] [Indexed: 11/20/2022]
Abstract
In vertebrates, lens epithelial cells cover the anterior half of the lens fiber core. During development, lens epithelial cells proliferate, move posteriorly and differentiate into lens fiber cells after passing through the equator. To elucidate the mechanisms underlying lens epithelial cell movement, we conducted time-lapse imaging of zebrafish lens epithelium. Lens epithelial cells do not intermingle but maintain their relative positions during development. Cell division induces epithelial rearrangement, which subsequently promotes cell movement towards the equator. These data suggest that cell division is the major driving force for cell movement. In zebrafish, E-cadherin is expressed in lens epithelium, whereas N-cadherin is required for lens fiber growth. E-cadherin reduced lens epithelial cell movement, whereas N-cadherin enhanced it. Laser ablation experiments revealed that lens epithelium is governed by pulling tension, which is modulated by these cadherins. Thus, cell division and cadherin-mediated adhesion regulate lens epithelial cell movement via modulation of epithelial tension.
Collapse
Affiliation(s)
- Toshiaki Mochizuki
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Yi-Jyun Luo
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Hsieh-Fu Tsai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Akane Hagiwara
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
33
|
The Molecular Basis of Radial Intercalation during Tissue Spreading in Early Development. Dev Cell 2017; 37:213-25. [PMID: 27165554 PMCID: PMC4865533 DOI: 10.1016/j.devcel.2016.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/18/2016] [Accepted: 04/08/2016] [Indexed: 02/08/2023]
Abstract
Radial intercalation is a fundamental process responsible for the thinning of multilayered tissues during large-scale morphogenesis; however, its molecular mechanism has remained elusive. Using amphibian epiboly, the thinning and spreading of the animal hemisphere during gastrulation, here we provide evidence that radial intercalation is driven by chemotaxis of cells toward the external layer of the tissue. This role of chemotaxis in tissue spreading and thinning is unlike its typical role associated with large-distance directional movement of cells. We identify the chemoattractant as the complement component C3a, a factor normally linked with the immune system. The mechanism is explored by computational modeling and tested in vivo, ex vivo, and in vitro. This mechanism is robust against fluctuations of chemoattractant levels and expression patterns and explains expansion during epiboly. This study provides insight into the fundamental process of radial intercalation and could be applied to a wide range of morphogenetic events.
Collapse
|
34
|
Abstract
Ongoing work shows that misplaced epithelial cells have the capacity to reintegrate back into tissue layers. This movement appears to underlie tissue stability and may also control aspects of tissue structure. A recent study reveals that cell reintegration in at least one tissue, the Drosophila follicular epithelium, is based on adhesion molecules that line lateral cell surfaces. In this article we will review these observations, discuss their implications for epithelial tissue development and maintenance, and identify future directions for study.
Collapse
Affiliation(s)
- Tyler J Wilson
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Dan T Bergstralh
- Department of Biology, University of Rochester, Rochester, NY, USA
| |
Collapse
|
35
|
Smutny M, Ákos Z, Grigolon S, Shamipour S, Ruprecht V, Čapek D, Behrndt M, Papusheva E, Tada M, Hof B, Vicsek T, Salbreux G, Heisenberg CP. Friction forces position the neural anlage. Nat Cell Biol 2017; 19:306-317. [PMID: 28346437 PMCID: PMC5635970 DOI: 10.1038/ncb3492] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/17/2017] [Indexed: 12/23/2022]
Abstract
During embryonic development, mechanical forces are essential for cellular rearrangements driving tissue morphogenesis. Here, we show that in the early zebrafish embryo, friction forces are generated at the interface between anterior axial mesoderm (prechordal plate, ppl) progenitors migrating towards the animal pole and neurectoderm progenitors moving in the opposite direction towards the vegetal pole of the embryo. These friction forces lead to global rearrangement of cells within the neurectoderm and determine the position of the neural anlage. Using a combination of experiments and simulations, we show that this process depends on hydrodynamic coupling between neurectoderm and ppl as a result of E-cadherin-mediated adhesion between those tissues. Our data thus establish the emergence of friction forces at the interface between moving tissues as a critical force-generating process shaping the embryo.
Collapse
Affiliation(s)
- Michael Smutny
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Zsuzsa Ákos
- Department of Biological Physics, Eötvös
University, Pázmány Péter sétány 1A, Budapest
H-1117, Hungary
| | - Silvia Grigolon
- The Francis Crick Institute, 1 Midland Road, London NW1
1AT, UK
| | - Shayan Shamipour
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Verena Ruprecht
- Centre for Genomic Regulation (CRG), The Barcelona
Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003, Barcelona,
Spain
| | - Daniel Čapek
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Martin Behrndt
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Ekaterina Papusheva
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Masazumi Tada
- Department of Cell and Developmental Biology, University
College London, Gower Street, London, WC1E 6BT, UK
| | - Björn Hof
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Tamás Vicsek
- Department of Biological Physics, Eötvös
University, Pázmány Péter sétány 1A, Budapest
H-1117, Hungary
| | | | | |
Collapse
|
36
|
Morris EAR, Bodin S, Delaval B, Comunale F, Georget V, Costa ML, Lutfalla G, Gauthier-Rouvière C. Flotillins control zebrafish epiboly through their role in cadherin-mediated cell-cell adhesion. Biol Cell 2017; 109:210-221. [DOI: 10.1111/boc.201700001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Eduardo A. Rios Morris
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
- MRI-CRBM Imaging facility, CNRS, UMR 5237; Université de Montpellier; Montpellier 34293 France
- Laboratório de Diferenciação Muscular e Citoesqueleto; Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ 21949-590 Brazil
| | - Stéphane Bodin
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Bénédicte Delaval
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Franck Comunale
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Virginie Georget
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
- MRI-CRBM Imaging facility, CNRS, UMR 5237; Université de Montpellier; Montpellier 34293 France
| | - Manoel L. Costa
- Laboratório de Diferenciação Muscular e Citoesqueleto; Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ 21949-590 Brazil
| | - Georges Lutfalla
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques; Université de Montpellier II et I; CNRS, UMR 5235 Montpellier 34095 France
| | - Cécile Gauthier-Rouvière
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| |
Collapse
|
37
|
Kozlovskaja-Gumbrienė A, Yi R, Alexander R, Aman A, Jiskra R, Nagelberg D, Knaut H, McClain M, Piotrowski T. Proliferation-independent regulation of organ size by Fgf/Notch signaling. eLife 2017; 6. [PMID: 28085667 PMCID: PMC5235355 DOI: 10.7554/elife.21049] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/23/2016] [Indexed: 12/31/2022] Open
Abstract
Organ morphogenesis depends on the precise orchestration of cell migration, cell shape changes and cell adhesion. We demonstrate that Notch signaling is an integral part of the Wnt and Fgf signaling feedback loop coordinating cell migration and the self-organization of rosette-shaped sensory organs in the zebrafish lateral line system. We show that Notch signaling acts downstream of Fgf signaling to not only inhibit hair cell differentiation but also to induce and maintain stable epithelial rosettes. Ectopic Notch expression causes a significant increase in organ size independently of proliferation and the Hippo pathway. Transplantation and RNASeq analyses revealed that Notch signaling induces apical junctional complex genes that regulate cell adhesion and apical constriction. Our analysis also demonstrates that in the absence of patterning cues normally provided by a Wnt/Fgf signaling system, rosettes still self-organize in the presence of Notch signaling. DOI:http://dx.doi.org/10.7554/eLife.21049.001
Collapse
Affiliation(s)
| | - Ren Yi
- Stowers Institute for Medical Research, Kansas City, United States
| | | | - Andy Aman
- Stowers Institute for Medical Research, Kansas City, United States
| | - Ryan Jiskra
- Stowers Institute for Medical Research, Kansas City, United States
| | - Danielle Nagelberg
- Developmental Genetics Program and Kimmel Center for Stem Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, United States
| | - Holger Knaut
- Developmental Genetics Program and Kimmel Center for Stem Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, United States
| | - Melainia McClain
- Stowers Institute for Medical Research, Kansas City, United States
| | | |
Collapse
|
38
|
Izaguirre MF, Casco VH. E-cadherin roles in animal biology: A perspective on thyroid hormone-influence. Cell Commun Signal 2016; 14:27. [PMID: 27814736 PMCID: PMC5097364 DOI: 10.1186/s12964-016-0150-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/26/2016] [Indexed: 01/15/2023] Open
Abstract
The establishment, remodeling and maintenance of tissular architecture during animal development, and even across juvenile to adult life, are deeply regulated by a delicate interplay of extracellular signals, cell membrane receptors and intracellular signal messengers. It is well known that cell adhesion molecules (cell-cell and cell-extracellular matrix) play a critical role in these processes. Particularly, adherens junctions (AJs) mediated by E-cadherin and catenins determine cell-cell contact survival and epithelia function. Consequently, this review seeks to encompass the complex and prolific knowledge about E-cadherin roles during physiological and pathological states, particularly focusing on the influence exerted by the thyroid hormone (TH).
Collapse
Affiliation(s)
- María Fernanda Izaguirre
- Laboratorio de Microscopia Aplicada a Estudios Moleculares y Celulares, Facultad de Ingeniería (Bioingeniería-Bioinformática), Universidad Nacional de Entre Ríos, Ruta 11, Km 10, Oro Verde, Entre Ríos, Argentina
| | - Victor Hugo Casco
- Laboratorio de Microscopia Aplicada a Estudios Moleculares y Celulares, Facultad de Ingeniería (Bioingeniería-Bioinformática), Universidad Nacional de Entre Ríos, Ruta 11, Km 10, Oro Verde, Entre Ríos, Argentina.
| |
Collapse
|
39
|
Han MKL, Hoijman E, Nöel E, Garric L, Bakkers J, de Rooij J. αE-catenin-dependent mechanotransduction is essential for proper convergent extension in zebrafish. Biol Open 2016; 5:1461-1472. [PMID: 27612508 PMCID: PMC5087688 DOI: 10.1242/bio.021378] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cadherin complexes mediate cell-cell adhesion and are crucial for embryonic development. Besides their structural function, cadherin complexes also transduce tension across the junction-actomyosin axis into proportional biochemical responses. Central to this mechanotransduction is the stretching of the cadherin-F-actin-linker α-catenin, which opens its central domain for binding to effectors such as vinculin. Mechanical unfolding of α-catenin leads to force-dependent reinforcement of cadherin-based junctions as studied in cell culture. The importance of cadherin mechanotransduction for embryonic development has not been studied yet. Here we used TALEN-mediated gene disruption to perturb endogenous αE-catenin in zebrafish development. Zygotic α-catenin mutants fail to maintain their epithelial barrier, resulting in tissue rupturing. We then specifically disrupted mechanotransduction, while maintaining cadherin adhesion, by expressing an αE-catenin construct in which the mechanosensitive domain was perturbed. Expression of either wild-type or mechano-defective α-catenin fully rescues barrier function in α-catenin mutants; however, expression of mechano-defective α-catenin also induces convergence and extension defects. Specifically, the polarization of cadherin-dependent, lamellipodia-driven cell migration of the lateral mesoderm was lost. These results indicate that cadherin mechanotransduction is crucial for proper zebrafish morphogenesis, and uncover one of the essential processes affected by its perturbation. Summary: Cadherin adhesions transduce tension across the junction-actomyosin axis into proportional biochemical responses via α-catenin. Here we show for the first time how this function of the cadherin complex is important during zebrafish morphogenesis.
Collapse
Affiliation(s)
- Mitchell K L Han
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht 3584CG, The Netherlands
| | - Esteban Hoijman
- The Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center Utrecht, Uppsalalaan 8, Utrecht 3584CT, The Netherlands
| | - Emily Nöel
- The Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center Utrecht, Uppsalalaan 8, Utrecht 3584CT, The Netherlands
| | - Laurence Garric
- The Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center Utrecht, Uppsalalaan 8, Utrecht 3584CT, The Netherlands
| | - Jeroen Bakkers
- The Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Center Utrecht, Uppsalalaan 8, Utrecht 3584CT, The Netherlands Department of Medical Physiology, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584 CM, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht 3584CG, The Netherlands
| |
Collapse
|
40
|
Zinc transporter ZIP10 forms a heteromer with ZIP6 which regulates embryonic development and cell migration. Biochem J 2016; 473:2531-44. [PMID: 27274087 PMCID: PMC4980808 DOI: 10.1042/bcj20160388] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/06/2016] [Indexed: 12/18/2022]
Abstract
Zinc is involved in cell migration during embryo development and in cancer. We show that a zinc transporter consisting of two proteins, ZIP6 and ZIP10, stimulates both cell migration and division in mammalian cells and in the zebrafish embryo. There is growing evidence that zinc and its transporters are involved in cell migration during development and in cancer. In the present study, we show that zinc transporter ZIP10 (SLC39A10) stimulates cell motility and proliferation, both in mammalian cells and in the zebrafish embryo. This is associated with inactivation of GSK (glycogen synthase kinase)-3α and -3β and down-regulation of E-cadherin (CDH1). Morpholino-mediated knockdown of zip10 causes delayed epiboly and deformities of the head, eye, heart and tail. Furthermore, zip10 deficiency results in overexpression of cdh1, zip6 and stat3, the latter gene product driving transcription of both zip6 and zip10. The non-redundant requirement of Zip6 and Zip10 for epithelial to mesenchymal transition (EMT) is consistent with our finding that they exist as a heteromer. We postulate that a subset of ZIPs carrying prion protein (PrP)-like ectodomains, including ZIP6 and ZIP10, are integral to cellular pathways and plasticity programmes, such as EMT.
Collapse
|
41
|
Abstract
Embryonic morphogenesis takes place via a series of dramatic collective cell movements. The mechanisms that coordinate these intricate structural transformations across an entire organism are not well understood. In this study, we used gentle mechanical deformation of developing zebrafish embryos to probe the role of physical forces in generating long-range intercellular coordination during epiboly, the process in which the blastoderm spreads over the yolk cell. Geometric distortion of the embryo resulted in nonuniform blastoderm migration and realignment of the anterior-posterior (AP) axis, as defined by the locations at which the head and tail form, toward the new long axis of the embryo and away from the initial animal-vegetal axis defined by the starting location of the blastoderm. We found that local alterations in the rate of blastoderm migration correlated with the local geometry of the embryo. Chemical disruption of the contractile ring of actin and myosin immediately vegetal to the blastoderm margin via Ca2+ reduction or treatment with blebbistatin restored uniform migration and eliminated AP axis reorientation in mechanically deformed embryos; it also resulted in cellular disorganization at the blastoderm margin. Our results support a model in which tension generated by the contractile actomyosin ring coordinates epiboly on both the organismal and cellular scales. Our observations likewise suggest that the AP axis is distinct from the initial animal-vegetal axis in zebrafish.
Collapse
|
42
|
Sempou E, Biasini E, Pinzón-Olejua A, Harris DA, Málaga-Trillo E. Activation of zebrafish Src family kinases by the prion protein is an amyloid-β-sensitive signal that prevents the endocytosis and degradation of E-cadherin/β-catenin complexes in vivo. Mol Neurodegener 2016; 11:18. [PMID: 26860872 PMCID: PMC4748561 DOI: 10.1186/s13024-016-0076-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/18/2016] [Indexed: 11/25/2022] Open
Abstract
Background Prions and amyloid-β (Aβ) oligomers trigger neurodegeneration by hijacking a poorly understood cellular signal mediated by the prion protein (PrP) at the plasma membrane. In early zebrafish embryos, PrP-1-dependent signals control cell-cell adhesion via a tyrosine phosphorylation-dependent mechanism. Results Here we report that the Src family kinases (SFKs) Fyn and Yes act downstream of PrP-1 to prevent the endocytosis and degradation of E-cadherin/β-catenin adhesion complexes in vivo. Accordingly, knockdown of PrP-1 or Fyn/Yes cause similar zebrafish gastrulation phenotypes, whereas Fyn/Yes expression rescues the PrP-1 knockdown phenotype. We also show that zebrafish and mouse PrPs positively regulate the activity of Src kinases and that these have an unexpected positive effect on E-cadherin-mediated cell adhesion. Interestingly, while PrP knockdown impairs β-catenin adhesive function, PrP overexpression enhances it, thereby antagonizing its nuclear, wnt-related signaling activity and disturbing embryonic dorsoventral specification. The ability of mouse PrP to influence these events in zebrafish embryos requires its neuroprotective, polybasic N-terminus but not its neurotoxicity-associated central region. Remarkably, human Aβ oligomers up-regulate the PrP-1/SFK/E-cadherin/β-catenin pathway in zebrafish embryonic cells, mimicking a PrP gain-of-function scenario. Conclusions Our gain- and loss-of-function experiments in zebrafish suggest that PrP and SFKs enhance the cell surface stability of embryonic adherens junctions via the same complex mechanism through which they over-activate neuroreceptors that trigger synaptic damage. The profound impact of this pathway on early zebrafish development makes these embryos an ideal model to study the cellular and molecular events affected by neurotoxic PrP mutations and ligands in vivo. In particular, our finding that human Aβ oligomers activate the zebrafish PrP/SFK/E-cadherin pathway opens the possibility of using fish embryos to rapidly screen for novel therapeutic targets and compounds against prion- and Alzheimer's-related neurodegeneration. Altogether, our data illustrate PrP-dependent signals relevant to embryonic development, neuronal physiology and neurological disease. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0076-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emily Sempou
- Department of Biology, University of Konstanz, Constance, 78457, Germany. .,Present address: Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Emiliano Biasini
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA. .,Present address: Dulbecco Telethon Institute, Laboratory of Prions and Amyloids, Centre for Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy.
| | - Alejandro Pinzón-Olejua
- Department of Biology, University of Konstanz, Constance, 78457, Germany. .,Present address: Max PIanck Institute for Brain Research, Department of Synaptic Plasticity, 60438, Frankfurt/Main, Germany.
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Edward Málaga-Trillo
- Department of Biology, University of Konstanz, Constance, 78457, Germany. .,Department of Biology, Universidad Peruana Cayetano Heredia, Lima 31, Perú.
| |
Collapse
|
43
|
Kent B, Magnani E, Walsh MJ, Sadler KC. UHRF1 regulation of Dnmt1 is required for pre-gastrula zebrafish development. Dev Biol 2016; 412:99-113. [PMID: 26851214 DOI: 10.1016/j.ydbio.2016.01.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 01/22/2016] [Accepted: 01/27/2016] [Indexed: 12/16/2022]
Abstract
Landmark epigenetic events underlie early embryonic development, yet how epigenetic modifiers are regulated to achieve rapid epigenome re-patterning is not known. Uhrf1 and DNA methyltransferase 1 (Dnmt1) are known to largely mediate maintenance DNA methylation and Uhrf1 is also required for both Dnmt1 localization and stability. Here, we investigate how these two key epigenetic modifiers regulate early zebrafish development and characterize the developmental consequences of disrupting their homeostatic relationship. Unlike Uhrf1 knockdown, which causes developmental arrest and death prior to gastrulation, overexpression of human UHRF1 (WT-UHRF1) caused asymmetric epiboly, inefficient gastrulation and multi-systemic defects. UHRF1 phosphorylation was previously demonstrated as essential for zebrafish embryogenesis, and we found that penetrance of the asymmetric epiboly phenotype was significantly increased in embryos injected with mRNA encoding non-phosphorylatable UHRF1 (UHRF1(S661A)). Surprisingly, both WT-UHRF1 and UHRF1(S661A) overexpression caused DNA hypomethylation. However, since other approaches that caused an equivalent degree of DNA hypomethylation did not cause the asymmetric epiboly phenotype, we conclude that bulk DNA methylation is not the primary mechanism. Instead, UHRF1(S661A) overexpression resulted in accumulation of Dnmt1 protein and the overexpression of both WT and a catalytically inactive Dnmt1 phenocopied the assymetric epiboly phenotype. Dnmt1 knockdown suppressed the phenotype caused by UHRF1(S661A) overexpression, and Uhrf1 knockdown suppressed the effect of Dnmt1 overexpression. Therefore, we conclude that the interaction between these two proteins is the mechanism underlying the gastrulation defects. This indicates that Dnmt1 stability requires UHRF1 phosphorylation and that crosstalk between the proteins is essential for the function of these two important epigenetic regulators during gastrulation.
Collapse
Affiliation(s)
- Brandon Kent
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States
| | - Elena Magnani
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States; Department of Medicine/Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States
| | - Martin J Walsh
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States; Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States; Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States
| | - Kirsten C Sadler
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States; Department of Medicine/Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, Box 1020, 1 Gustave L. Levy Place, New York, NY 10029, United States.
| |
Collapse
|
44
|
Bruce AE. Zebrafish epiboly: Spreading thin over the yolk. Dev Dyn 2015; 245:244-58. [DOI: 10.1002/dvdy.24353] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 01/07/2023] Open
Affiliation(s)
- Ashley E.E. Bruce
- Department of Cell and Systems Biology; University of Toronto; Toronto ON Canada
| |
Collapse
|
45
|
Matsui T, Ishikawa H, Bessho Y. Cell collectivity regulation within migrating cell cluster during Kupffer's vesicle formation in zebrafish. Front Cell Dev Biol 2015; 3:27. [PMID: 26000276 PMCID: PMC4423447 DOI: 10.3389/fcell.2015.00027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/22/2015] [Indexed: 02/04/2023] Open
Abstract
Although cell adhesion is thought to fasten cells tightly, cells that adhere to each other can migrate directionally. This group behavior, called “collective cell migration,” is observed during normal development, wound healing, and cancer invasion. Loss-of-function of cell adhesion molecules in several model systems of collective cell migration results in delay or inhibition of migration of cell groups but does not lead to dissociation of the cell groups, suggesting that mechanisms of cells staying assembled as a single cell cluster, termed as “cell collectivity,” remain largely unknown. During the formation of Kupffer's vesicle (KV, an organ of laterality in zebrafish), KV progenitors form a cluster and migrate together toward the vegetal pole. Importantly, in this model system of collective cell migration, knockdown of cell adhesion molecules or signal components leads to failure of cell collectivity. In this review, we summarize recent findings in cell collectivity regulation during collective migration of KV progenitor cells and describe our current understanding of how cell collectivity is regulated during collective cell migration.
Collapse
Affiliation(s)
- Takaaki Matsui
- Gene Regulation Research, Nara Institute of Science and Technology Nara, Japan
| | - Hiroshi Ishikawa
- Gene Regulation Research, Nara Institute of Science and Technology Nara, Japan
| | - Yasumasa Bessho
- Gene Regulation Research, Nara Institute of Science and Technology Nara, Japan
| |
Collapse
|
46
|
Xiong F, Ma W, Hiscock TW, Mosaliganti KR, Tentner AR, Brakke KA, Rannou N, Gelas A, Souhait L, Swinburne IA, Obholzer ND, Megason SG. Interplay of cell shape and division orientation promotes robust morphogenesis of developing epithelia. Cell 2015; 159:415-27. [PMID: 25303534 DOI: 10.1016/j.cell.2014.09.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/29/2014] [Accepted: 09/03/2014] [Indexed: 11/17/2022]
Abstract
Epithelial cells acquire functionally important shapes (e.g., squamous, cuboidal, columnar) during development. Here, we combine theory, quantitative imaging, and perturbations to analyze how tissue geometry, cell divisions, and mechanics interact to shape the presumptive enveloping layer (pre-EVL) on the zebrafish embryonic surface. We find that, under geometrical constraints, pre-EVL flattening is regulated by surface cell number changes following differentially oriented cell divisions. The division pattern is, in turn, determined by the cell shape distribution, which forms under geometrical constraints by cell-cell mechanical coupling. An integrated mathematical model of this shape-division feedback loop recapitulates empirical observations. Surprisingly, the model predicts that cell shape is robust to changes of tissue surface area, cell volume, and cell number, which we confirm in vivo. Further simulations and perturbations suggest the parameter linking cell shape and division orientation contributes to epithelial diversity. Together, our work identifies an evolvable design logic that enables robust cell-level regulation of tissue-level development.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenzhe Ma
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom W Hiscock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrea R Tentner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth A Brakke
- Mathematics Department, Susquehanna University, Selinsgrove, PA 17870, USA
| | - Nicolas Rannou
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Gelas
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lydie Souhait
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ian A Swinburne
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nikolaus D Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Lima A, Cha BJ, Amin J, Smith LK, Anderson B. Zebrafish embryo model of Bartonella henselae infection. Zebrafish 2014; 11:434-46. [PMID: 25026365 DOI: 10.1089/zeb.2014.1001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bartonella henselae (Bh) is an emerging zoonotic pathogen that has been associated with a variety of human diseases, including bacillary angiomatosis that is characterized by vasoproliferative tumor-like lesions on the skin of some immunosuppressed individuals. The study of Bh pathogenesis has been limited to in vitro cell culture systems due to the lack of an animal model. Therefore, we wanted to investigate whether the zebrafish embryo could be used to model human infection with Bh. Our data showed that Tg(fli1:egfp)(y1) zebrafish embryos supported a sustained Bh infection for 7 days with >10-fold bacterial replication when inoculated in the yolk sac. We showed that Bh recruited phagocytes to the site of infection in the Tg(mpx:GFP)uwm1 embryos. Infected embryos showed evidence of a Bh-induced angiogenic phenotype and an increase in the expression of genes encoding pro-inflammatory factors and pro-angiogenic markers. However, infection of zebrafish embryos with a deletion mutant in the major adhesin (BadA) resulted in little or no bacterial replication and a diminished host response, providing the first evidence that BadA is critical for in vivo infection. Thus, the zebrafish embryo provides the first practical model of Bh infection that will facilitate efforts to identify virulence factors and define molecular mechanisms of Bh pathogenesis.
Collapse
Affiliation(s)
- Amorce Lima
- 1 Department of Molecular Medicine, University of South Florida Morsani College of Medicine , Tampa, Florida
| | | | | | | | | |
Collapse
|
48
|
Cell adhesion in zebrafish embryos is modulated by March 8. PLoS One 2014; 9:e94873. [PMID: 24752240 PMCID: PMC3994051 DOI: 10.1371/journal.pone.0094873] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 03/20/2014] [Indexed: 12/16/2022] Open
Abstract
March8 is a member of a family of transmembrane E3 ubiquitin ligases that have been studied mostly for their role in the immune system. We find that March8 is expressed in the zebrafish egg and early embryo, suggesting a role in development. Both knock-down and overexpression of March8 leads to abnormal development. The phenotype of zebrafish embryos and Xenopus animal explants overexpressing March8 implicates impairment of cell adhesion as a cause of the effect. In zebrafish embryos and in cultured cells, overexpression of March8 leads to a reduction in the surface levels of E-cadherin, a major cell-cell adhesion molecule. Experiments in cell culture further show that E-cadherin can be ubiquitinated by March8. On the basis of these observations we suggest that March8 functions in the embryo to modulate the strength of cell adhesion by regulating the localization of E-cadherin.
Collapse
|
49
|
Li C, Ma H, Wang Y, Cao Z, Graves-Deal R, Powell AE, Starchenko A, Ayers GD, Washington MK, Kamath V, Desai K, Gerdes MJ, Solnica-Krezel L, Coffey RJ. Excess PLAC8 promotes an unconventional ERK2-dependent EMT in colon cancer. J Clin Invest 2014; 124:2172-87. [PMID: 24691442 DOI: 10.1172/jci71103] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 01/30/2014] [Indexed: 01/30/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) transcriptional program is characterized by repression of E-cadherin (CDH1) and induction of N-cadherin (CDH2), and mesenchymal genes like vimentin (VIM). Placenta-specific 8 (PLAC8) has been implicated in colon cancer; however, how PLAC8 contributes to disease is unknown, and endogenous PLAC8 protein has not been studied. We analyzed zebrafish and human tissues and found that endogenous PLAC8 localizes to the apical domain of differentiated intestinal epithelium. Colon cancer cells with elevated PLAC8 levels exhibited EMT features, including increased expression of VIM and zinc finger E-box binding homeobox 1 (ZEB1), aberrant cell motility, and increased invasiveness. In contrast to classical EMT, PLAC8 overexpression reduced cell surface CDH1 and upregulated P-cadherin (CDH3) without affecting CDH2 expression. PLAC8-induced EMT was linked to increased phosphorylated ERK2 (p-ERK2), and ERK2 knockdown restored cell surface CDH1 and suppressed CDH3, VIM, and ZEB1 upregulation. In vitro, PLAC8 directly bound and inactivated the ERK2 phosphatase DUSP6, thereby increasing p-ERK2. In a murine xenograft model, knockdown of endogenous PLAC8 in colon cancer cells resulted in smaller tumors, reduced local invasion, and decreased p-ERK2. Using MultiOmyx, a multiplex immunofluorescence-based methodology, we observed coexpression of cytosolic PLAC8, CDH3, and VIM at the leading edge of a human colorectal tumor, supporting a role for PLAC8 in cancer invasion in vivo.
Collapse
|
50
|
Planar cell polarity proteins differentially regulate extracellular matrix organization and assembly during zebrafish gastrulation. Dev Biol 2013; 383:39-51. [DOI: 10.1016/j.ydbio.2013.08.027] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/25/2013] [Accepted: 08/31/2013] [Indexed: 11/22/2022]
|