1
|
Chang CT, Tsai T, Solnica-Krezel L. Temporal regulation of endoderm convergence and extension by the BMP activity gradient through mesoderm-dependent and independent mechanisms. Cells Dev 2025:204021. [PMID: 40090551 DOI: 10.1016/j.cdev.2025.204021] [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: 12/30/2024] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
One hundred years ago, Spemann and Mangold identified the organizer, a critical embryonic region that establishes vertebrate body axes by directing cell fate and morphogenesis. A conserved vertebrate mechanism involves the regulation of a ventral-to-dorsal BMP activity gradient during gastrulation by the organizer-expressed molecules. In zebrafish, BMP signaling controls mesodermal cell convergence and extension (C&E) by inhibiting Planar Cell Polarity (PCP) signaling and regulating cell adhesion. This allows lateral cells to converge toward the dorsal midline while directing ventral cells toward the tail bud. However, BMP's role in endodermal cell movements and the temporal precision of its regulatory functions remain poorly understood. Using optogenetics and other loss- and gain-of-function approaches, we investigated BMP's role in mesoderm and endoderm C&E. We found that low BMP signaling promotes extension in both germ layers, whereas high BMP signaling inhibits their C&E. Remarkably, BMP signaling activation for 1 h rapidly redirected dorsal to ventral migration of both mesodermal and endodermal cells. However, when BMP signaling was selectively elevated in endoderm in embryos with reduced BMP signaling, endoderm still mimicked mesodermal cell movements, indicating that endodermal responses to BMP are non-cell autonomous. We show that movements of endodermal cells in gastrulae with normal or elevated BMP signaling are not entirely dependent on mesoderm or the Cxcl12b/Cxcr4a GPCR pathway, suggesting additional mechanisms underlie endoderm C&E. Our findings highlight the critical role of the BMP morphogen gradient in coordinated C&E movements of mesodermal and endodermal cells. BMP employs both direct and indirect mechanisms to ensure robust embryonic patterning and morphogenesis of germ layers.
Collapse
Affiliation(s)
- Chia-Teng Chang
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, MO 63110, USA
| | - Tony Tsai
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, MO 63110, USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Johal S, Elsayed R, Panfilio KA, Nelson AC. The molecular basis for functional divergence of duplicated SOX factors controlling endoderm formation and left-right patterning in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579092. [PMID: 39605568 PMCID: PMC11601245 DOI: 10.1101/2024.02.06.579092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Endoderm, one of three primary germ layers of vertebrate embryos, makes major contributions to the respiratory and gastrointestinal tracts and associated organs, including liver and pancreas. In mammals, the transcription factor SOX17 is vital for endoderm organ formation and can induce endoderm progenitor identity. Duplication of ancestral sox17 in the teleost lineage produced the paralogues sox32 and sox17 in zebrafish. Sox32 is required for specification of endoderm and progenitors of the left-right organiser (Kupffer's Vesicle, KV), with Sox17 a downstream target of Sox32 that is implicated in further KV development. Phenotypic evidence therefore suggests functional similarities between zebrafish Sox32 and Sox17 and mammalian SOX17. Here, we directly compare these orthologues and paralogues, using the early zebrafish embryo as a biological platform for functional testing. Our results indicate that, unlike Sox32, human SOX17 cannot induce endoderm specification in zebrafish. Furthermore, using hybrid protein functional analyses, we show that Sox32 specificity for the endoderm gene regulatory network is linked to evolutionary divergence in its DNA-binding HMG domain from its paralogue Sox17. Additionally, changes in the C-terminal regions of Sox32 and Sox17 underpin their differing target specificities. Finally, we establish that specific conserved peptides in the C-terminal domain are essential for the role of Sox17 in establishing correct organ asymmetry. Overall, our results illuminate the molecular basis for functional divergence of Sox32 and Sox17 in vertebrate endoderm development and left-right patterning, and the evolution of SoxF transcription factor function.
Collapse
|
3
|
Moran HR, Nyarko OO, O’Rourke R, Ching RCK, Riemslagh FW, Peña B, Burger A, Sucharov CC, Mosimann C. The pericardium forms as a distinct structure during heart formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613484. [PMID: 39345600 PMCID: PMC11429720 DOI: 10.1101/2024.09.18.613484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The heart integrates diverse cell lineages into a functional unit, including the pericardium, a mesothelial sac that supports heart movement, homeostasis, and immune responses. However, despite its critical roles, the developmental origins of the pericardium remain uncertain due to disparate models. Here, using live imaging, lineage tracking, and single-cell transcriptomics in zebrafish, we find the pericardium forms within the lateral plate mesoderm from dedicated anterior mesothelial progenitors and distinct from the classic heart field. Imaging of transgenic reporters in zebrafish documents lateral plate mesoderm cells that emerge lateral of the classic heart field and among a continuous mesothelial progenitor field. Single-cell transcriptomics and trajectories of hand2-expressing lateral plate mesoderm reveal distinct populations of mesothelial and cardiac precursors, including pericardial precursors that are distinct from the cardiomyocyte lineage. The mesothelial gene expression signature is conserved in mammals and carries over to post-natal development. Light sheet-based live-imaging and machine learning-supported cell tracking documents that during heart tube formation, pericardial precursors that reside at the anterior edge of the heart field migrate anteriorly and medially before fusing, enclosing the embryonic heart to form a single pericardial cavity. Pericardium formation proceeds even upon genetic disruption of heart tube formation, uncoupling the two structures. Canonical Wnt/β-catenin signaling modulates pericardial cell number, resulting in a stretched pericardial epithelium with reduced cell number upon canonical Wnt inhibition. We connect the pathological expression of secreted Wnt antagonists of the SFRP family found in pediatric dilated cardiomyopathy to increased pericardial stiffness: sFRP1 in the presence of increased catecholamines causes cardiomyocyte stiffness in neonatal rats as measured by atomic force microscopy. Altogether, our data integrate pericardium formation as an independent process into heart morphogenesis and connect disrupted pericardial tissue properties such as pericardial stiffness to pediatric cardiomyopathies.
Collapse
Affiliation(s)
- Hannah R. Moran
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Obed O. Nyarko
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ryenne-Christine K. Ching
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Frederike W. Riemslagh
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Institute, Division of Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Bioengineering Department, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
4
|
Kurup AJ, Bailet F, Fürthauer M. Myosin1G promotes Nodal signaling to control zebrafish left-right asymmetry. Nat Commun 2024; 15:6547. [PMID: 39095343 PMCID: PMC11297164 DOI: 10.1038/s41467-024-50868-y] [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: 09/03/2023] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Myosin1D (Myo1D) has recently emerged as a conserved regulator of animal Left-Right (LR) asymmetry that governs the morphogenesis of the vertebrate central LR Organizer (LRO). In addition to Myo1D, the zebrafish genome encodes the closely related Myo1G. Here we show that while Myo1G also controls LR asymmetry, it does so through an entirely different mechanism. Myo1G promotes the Nodal-mediated transfer of laterality information from the LRO to target tissues. At the cellular level, Myo1G is associated with endosomes positive for the TGFβ signaling adapter SARA. myo1g mutants have fewer SARA-positive Activin receptor endosomes and a reduced responsiveness to Nodal ligands that results in a delay of left-sided Nodal propagation and tissue-specific laterality defects in organs that are most distant from the LRO. Additionally, Myo1G promotes signaling by different Nodal ligands in specific biological contexts. Our findings therefore identify Myo1G as a context-dependent regulator of the Nodal signaling pathway.
Collapse
|
5
|
Noël ES. Cardiac construction-Recent advances in morphological and transcriptional modeling of early heart development. Curr Top Dev Biol 2024; 156:121-156. [PMID: 38556421 DOI: 10.1016/bs.ctdb.2024.02.005] [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] [Indexed: 04/02/2024]
Abstract
During human embryonic development the early establishment of a functional heart is vital to support the growing fetus. However, forming the embryonic heart is an extremely complex process, requiring spatiotemporally controlled cell specification and differentiation, tissue organization, and coordination of cardiac function. These complexities, in concert with the early and rapid development of the embryonic heart, mean that understanding the intricate interplay between these processes that help shape the early heart remains highly challenging. In this review I focus on recent insights from animal models that have shed new light on the earliest stages of heart development. This includes specification and organization of cardiac progenitors, cell and tissue movements that make and shape the early heart tube, and the initiation of the first beat in the developing heart. In addition I highlight relevant in vitro models that could support translation of findings from animal models to human heart development. Finally I discuss challenges that are being addressed in the field, along with future considerations that together may help move us towards a deeper understanding of how our hearts are made.
Collapse
Affiliation(s)
- Emily S Noël
- School of Biosciences and Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
6
|
Shrestha R, McCann T, Saravanan H, Lieberth J, Koirala P, Bloomekatz J. The myocardium utilizes a platelet-derived growth factor receptor alpha (Pdgfra)-phosphoinositide 3-kinase (PI3K) signaling cascade to steer toward the midline during zebrafish heart tube formation. eLife 2023; 12:e85930. [PMID: 37921445 PMCID: PMC10651176 DOI: 10.7554/elife.85930] [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: 01/13/2023] [Accepted: 11/02/2023] [Indexed: 11/04/2023] Open
Abstract
Coordinated cell movement is a fundamental process in organ formation. During heart development, bilateral myocardial precursors collectively move toward the midline (cardiac fusion) to form the primitive heart tube. Extrinsic influences such as the adjacent anterior endoderm are known to be required for cardiac fusion. We previously showed however, that the platelet-derived growth factor receptor alpha (Pdgfra) is also required for cardiac fusion (Bloomekatz et al., 2017). Nevertheless, an intrinsic mechanism that regulates myocardial movement has not been elucidated. Here, we show that the phosphoinositide 3-kinase (PI3K) intracellular signaling pathway has an essential intrinsic role in the myocardium directing movement toward the midline. In vivo imaging further reveals midline-oriented dynamic myocardial membrane protrusions that become unpolarized in PI3K-inhibited zebrafish embryos where myocardial movements are misdirected and slower. Moreover, we find that PI3K activity is dependent on and interacts with Pdgfra to regulate myocardial movement. Together our findings reveal an intrinsic myocardial steering mechanism that responds to extrinsic cues during the initiation of cardiac development.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of MississippiUniversityUnited States
| | - Tess McCann
- Department of Biology, University of MississippiUniversityUnited States
| | - Harini Saravanan
- Department of Biology, University of MississippiUniversityUnited States
| | - Jaret Lieberth
- Department of Biology, University of MississippiUniversityUnited States
| | - Prashanna Koirala
- Department of Biology, University of MississippiUniversityUnited States
| | - Joshua Bloomekatz
- Department of Biology, University of MississippiUniversityUnited States
| |
Collapse
|
7
|
Brown W, Wesalo J, Samanta S, Luo J, Caldwell SE, Tsang M, Deiters A. Genetically Encoded Aminocoumarin Lysine for Optical Control of Protein-Nucleotide Interactions in Zebrafish Embryos. ACS Chem Biol 2023; 18:1305-1314. [PMID: 37272594 PMCID: PMC10278064 DOI: 10.1021/acschembio.3c00028] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
The strategic placement of unnatural amino acids into the active site of kinases and phosphatases has allowed for the generation of photocaged signaling proteins that offer spatiotemporal control over activation of these pathways through precise light exposure. However, deploying this technology to study cell signaling in the context of embryo development has been limited. The promise of optical control is especially useful in the early stages of an embryo where development is driven by tightly orchestrated signaling events. Here, we demonstrate light-induced activation of Protein Kinase A and a RASopathy mutant of NRAS in the zebrafish embryo using a new light-activated amino acid. We applied this approach to gain insight into the roles of these proteins in gastrulation and heart development and forge a path for further investigation of RASopathy mutant proteins in animals.
Collapse
Affiliation(s)
- Wes Brown
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Joshua Wesalo
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Subhas Samanta
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Ji Luo
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Steven E. Caldwell
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Michael Tsang
- Department
of Developmental Biology, University of
Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
8
|
van Boxtel AL. Whole-Mount In Situ Hybridization for Detection of Migrating Zebrafish Endodermal Cells. Methods Mol Biol 2023; 2608:131-145. [PMID: 36653706 DOI: 10.1007/978-1-0716-2887-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
One of the most important events in early vertebrate development is the formation and positioning of the endoderm, the embryonic progenitor cell population that gives rise to the internal organs. Recent years have seen renewed interest in the mechanisms underlying the specification and migration of endodermal progenitor cells. The zebrafish is a well-established, accessible, and powerful model to study this cell population. Zebrafish endodermal cells are specified around 4 h after fertilization and subsequently migrate as evenly spaced single cells in a stereotypical manner in the next 6 h. Given the large numbers of fertilized eggs that can be obtained from a single breeding pair and the ease of chemical and genetic perturbations, the zebrafish is an excellent model to study mechanisms underlying endoderm specification and migration. An easy approach to visualizing and quantitating endodermal cells and their migratory routes is by whole-mount in situ hybridization (WISH) on fixed embryos, collected in time series. This chapter provides basic information on the organization and staging of the embryos, with an emphasis on the migrating endodermal cell population. In addition, optimized protocols for the isolation and fixation of staged embryos are provided as well as detailed probe synthesis and WISH protocols, specific for migrating endoderm. Finally, details are provided on how to approach these experiments quantitatively, and some common pitfalls are discussed.
Collapse
Affiliation(s)
- Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Shrestha R, McCann T, Saravanan H, Lieberth J, Koirala P, Bloomekatz J. The myocardium utilizes Pdgfra-PI3K signaling to steer towards the midline during heart tube formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522612. [PMID: 36712046 PMCID: PMC9881939 DOI: 10.1101/2023.01.03.522612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Coordinated cell movement is a fundamental process in organ formation. During heart development, bilateral myocardial precursors collectively move towards the midline (cardiac fusion) to form the primitive heart tube. Along with extrinsic influences such as the adjacent anterior endoderm which are known to be required for cardiac fusion, we previously showed that the platelet-derived growth factor receptor alpha (Pdgfra) is also required. However, an intrinsic mechanism that regulates myocardial movement remains to be elucidated. Here, we uncover an essential intrinsic role in the myocardium for the phosphoinositide 3-kinase (PI3K) intracellular signaling pathway in directing myocardial movement towards the midline. In vivo imaging reveals that in PI3K-inhibited zebrafish embryos myocardial movements are misdirected and slower, while midline-oriented dynamic myocardial membrane protrusions become unpolarized. Moreover, PI3K activity is dependent on and genetically interacts with Pdgfra to regulate myocardial movement. Together our findings reveal an intrinsic myocardial steering mechanism that responds to extrinsic cues during the initiation of cardiac development.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, University, MS 38677
| | - Tess McCann
- Department of Biology, University of Mississippi, University, MS 38677
| | - Harini Saravanan
- Department of Biology, University of Mississippi, University, MS 38677
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, University, MS 38677
| | - Prashanna Koirala
- Department of Biology, University of Mississippi, University, MS 38677
| | - Joshua Bloomekatz
- Department of Biology, University of Mississippi, University, MS 38677
| |
Collapse
|
10
|
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: 0.3] [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
|
11
|
Talbot CD, Walsh MD, Cutty SJ, Elsayed R, Vlachaki E, Bruce AEE, Wardle FC, Nelson AC. Eomes function is conserved between zebrafish and mouse and controls left-right organiser progenitor gene expression via interlocking feedforward loops. Front Cell Dev Biol 2022; 10:982477. [PMID: 36133924 PMCID: PMC9483813 DOI: 10.3389/fcell.2022.982477] [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: 06/30/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The T-box family transcription factor Eomesodermin (Eomes) is present in all vertebrates, with many key roles in the developing mammalian embryo and immune system. Homozygous Eomes mutant mouse embryos exhibit early lethality due to defects in both the embryonic mesendoderm and the extraembryonic trophoblast cell lineage. In contrast, zebrafish lacking the predominant Eomes homologue A (Eomesa) do not suffer complete lethality and can be maintained. This suggests fundamental differences in either the molecular function of Eomes orthologues or the molecular configuration of processes in which they participate. To explore these hypotheses we initially analysed the expression of distinct Eomes isoforms in various mouse cell types. Next we compared the functional capabilities of these murine isoforms to zebrafish Eomesa. These experiments provided no evidence for functional divergence. Next we examined the functions of zebrafish Eomesa and other T-box family members expressed in early development, as well as its paralogue Eomesb. Though Eomes is a member of the Tbr1 subfamily we found evidence for functional redundancy with the Tbx6 subfamily member Tbx16, known to be absent from eutherians. However, Tbx16 does not appear to synergise with Eomesa cofactors Mixl1 and Gata5. Finally, we analysed the ability of Eomesa and other T-box factors to induce zebrafish left-right organiser progenitors (known as dorsal forerunner cells) known to be positively regulated by vgll4l, a gene we had previously shown to be repressed by Eomesa. Here we demonstrate that Eomesa indirectly upregulates vgll4l expression via interlocking feedforward loops, suggesting a role in establishment of left-right asymmetry. Conversely, other T-box factors could not similarly induce left-right organiser progenitors. Overall these findings demonstrate conservation of Eomes molecular function and participation in similar processes, but differential requirements across evolution due to additional co-expressed T-box factors in teleosts, albeit with markedly different molecular capabilities. Our analyses also provide insights into the role of Eomesa in left-right organiser formation in zebrafish.
Collapse
Affiliation(s)
- Conor D. Talbot
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Mark D. Walsh
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Stephen J. Cutty
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Randa Elsayed
- Warwick Medical School, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Eirini Vlachaki
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Ashley E. E. Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Fiona C. Wardle
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Andrew C. Nelson
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
12
|
Ikeda T, Inamori K, Kawanishi T, Takeda H. Reemployment of Kupffer's vesicle cells into axial and paraxial mesoderm via transdifferentiation. Dev Growth Differ 2022; 64:163-177. [PMID: 35129208 DOI: 10.1111/dgd.12774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
Kupffer's vesicle (KV) in the teleost embryo is a fluid-filled vesicle surrounded by a layer of epithelial cells with rotating primary cilia. KV transiently acts as the left-right organizer and degenerates after the establishment of left-right asymmetric gene expression. Previous labelling experiments in zebrafish embryos indicated that descendants of KV-epithelial cells are incorporated into mesodermal tissues after the collapse of KV. However, the overall picture of their differentiation potency had been unclear due to the lack of suitable genetic tools and molecular analyses. In the present study, we established a novel zebrafish transgenic line with a promoter of dand5, in which all KV-epithelial cells and their descendants are specifically labelled until the larval stage. We found that KV-epithelial cells undergo epithelial-mesenchymal transition upon KV collapse and infiltrate into adjacent mesodermal progenitors, the presomitic mesoderm and chordoneural hinge. Once incorporated, the descendants of KV-epithelial cells expressed distinct mesodermal differentiation markers and contributed to the mature populations such as the axial muscles and notochordal sheath through normal developmental process. These results indicate that differentiated KV-epithelial cells possess unique plasticity in that they are reemployed into mesodermal lineages through transdifferentiation after they complete their initial role in KV.
Collapse
Affiliation(s)
- Takafumi Ikeda
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kiichi Inamori
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Toru Kawanishi
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takeda
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Stark BC, Gao Y, Sepich DS, Belk L, Culver MA, Hu B, Mekel M, Ferris W, Shin J, Solnica-Krezel L, Lin F, Cooper JA. CARMIL3 is important for cell migration and morphogenesis during early development in zebrafish. Dev Biol 2022; 481:148-159. [PMID: 34599906 PMCID: PMC8781030 DOI: 10.1016/j.ydbio.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023]
Abstract
Cell migration is important during early animal embryogenesis. Cell migration and cell shape are controlled by actin assembly and dynamics, which depend on capping proteins, including the barbed-end heterodimeric actin capping protein (CP). CP activity can be regulated by capping-protein-interacting (CPI) motif proteins, including CARMIL (capping protein Arp2/3 myosin-I linker) family proteins. Previous studies of CARMIL3, one of the three highly conserved CARMIL genes in vertebrates, have largely been limited to cells in culture. Towards understanding CARMIL function during embryogenesis in vivo, we analyzed zebrafish lines carrying mutations of carmil3. Maternal-zygotic mutants showed impaired endodermal migration during gastrulation, along with defects in dorsal forerunner cell (DFC) cluster formation, which affected the morphogenesis of Kupffer's vesicle (KV). Mutant KVs were smaller, contained fewer cells and displayed decreased numbers of cilia, leading to defects in left/right (L/R) patterning with variable penetrance and expressivity. The penetrance and expressivity of the KV phenotype in carmil3 mutants correlated well with the L/R heart positioning defect at the end of embryogenesis. This in vivo animal study of CARMIL3 reveals its new role during morphogenesis of the vertebrate embryo. This role involves migration of endodermal cells and DFCs, along with subsequent morphogenesis of the KV and L/R asymmetry.
Collapse
Affiliation(s)
- Benjamin C. Stark
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO
| | - Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Diane S. Sepich
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO
| | - Lakyn Belk
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Matthew A. Culver
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Marlene Mekel
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO
| | - Wyndham Ferris
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO
| | - Jimann Shin
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO,Corresponding authors. Email addresses for correspondence after publication: Fang Lin, ; Lilianna Solnica-Krezel, ; John Cooper,
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA.,Corresponding authors. Email addresses for correspondence after publication: Fang Lin, ; Lilianna Solnica-Krezel, ; John Cooper,
| | - John A. Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO,Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO,Corresponding authors. Email addresses for correspondence after publication: Fang Lin, ; Lilianna Solnica-Krezel, ; John Cooper,
| |
Collapse
|
14
|
Hu B, Rodriguez JJ, Kakkerla Balaraju A, Gao Y, Nguyen NT, Steen H, Suhaib S, Chen S, Lin F. Glypican 4 mediates Wnt transport between germ layers via signaling filopodia. J Cell Biol 2021; 220:212673. [PMID: 34591076 PMCID: PMC8488972 DOI: 10.1083/jcb.202009082] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 07/18/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022] Open
Abstract
Glypicans influence signaling pathways by regulating morphogen trafficking and reception. However, the underlying mechanisms in vertebrates are poorly understood. In zebrafish, Glypican 4 (Gpc4) is required for convergence and extension (C&E) of both the mesoderm and endoderm. Here, we show that transgenic expression of GFP-Gpc4 in the endoderm of gpc4 mutants rescued C&E defects in all germ layers. The rescue of mesoderm was likely mediated by Wnt5b and Wnt11f2 and depended on signaling filopodia rather than on cleavage of the Gpc4 GPI anchor. Gpc4 bound both Wnt5b and Wnt11f2 and regulated formation of the filopodia that transport Wnt5b and Wnt11f2 to neighboring cells. Moreover, this rescue was suppressed by blocking signaling filopodia that extend from endodermal cells. Thus, GFP-Gpc4–labeled protrusions that emanated from endodermal cells transported Wnt5b and Wnt11f2 to other germ layers, rescuing the C&E defects caused by a gpc4 deficiency. Our study reveals a new mechanism that could explain in vivo morphogen distribution involving Gpc4.
Collapse
Affiliation(s)
- Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Juan J Rodriguez
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Anurag Kakkerla Balaraju
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Nhan T Nguyen
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Heston Steen
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Saeb Suhaib
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Songhai Chen
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| |
Collapse
|
15
|
Bornhorst D, Abdelilah-Seyfried S. Strong as a Hippo's Heart: Biomechanical Hippo Signaling During Zebrafish Cardiac Development. Front Cell Dev Biol 2021; 9:731101. [PMID: 34422841 PMCID: PMC8375320 DOI: 10.3389/fcell.2021.731101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The heart is comprised of multiple tissues that contribute to its physiological functions. During development, the growth of myocardium and endocardium is coupled and morphogenetic processes within these separate tissue layers are integrated. Here, we discuss the roles of mechanosensitive Hippo signaling in growth and morphogenesis of the zebrafish heart. Hippo signaling is involved in defining numbers of cardiac progenitor cells derived from the secondary heart field, in restricting the growth of the epicardium, and in guiding trabeculation and outflow tract formation. Recent work also shows that myocardial chamber dimensions serve as a blueprint for Hippo signaling-dependent growth of the endocardium. Evidently, Hippo pathway components act at the crossroads of various signaling pathways involved in embryonic zebrafish heart development. Elucidating how biomechanical Hippo signaling guides heart morphogenesis has direct implications for our understanding of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Stem Cell Program, Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
16
|
Kemmler CL, Riemslagh FW, Moran HR, Mosimann C. From Stripes to a Beating Heart: Early Cardiac Development in Zebrafish. J Cardiovasc Dev Dis 2021; 8:17. [PMID: 33578943 PMCID: PMC7916704 DOI: 10.3390/jcdd8020017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
The heart is the first functional organ to form during vertebrate development. Congenital heart defects are the most common type of human birth defect, many originating as anomalies in early heart development. The zebrafish model provides an accessible vertebrate system to study early heart morphogenesis and to gain new insights into the mechanisms of congenital disease. Although composed of only two chambers compared with the four-chambered mammalian heart, the zebrafish heart integrates the core processes and cellular lineages central to cardiac development across vertebrates. The rapid, translucent development of zebrafish is amenable to in vivo imaging and genetic lineage tracing techniques, providing versatile tools to study heart field migration and myocardial progenitor addition and differentiation. Combining transgenic reporters with rapid genome engineering via CRISPR-Cas9 allows for functional testing of candidate genes associated with congenital heart defects and the discovery of molecular causes leading to observed phenotypes. Here, we summarize key insights gained through zebrafish studies into the early patterning of uncommitted lateral plate mesoderm into cardiac progenitors and their regulation. We review the central genetic mechanisms, available tools, and approaches for modeling congenital heart anomalies in the zebrafish as a representative vertebrate model.
Collapse
Affiliation(s)
| | | | | | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (C.L.K.); (F.W.R.); (H.R.M.)
| |
Collapse
|
17
|
Mao A, Zhang M, Li L, Liu J, Ning G, Cao Y, Wang Q. Pharyngeal pouches provide a niche microenvironment for arch artery progenitor specification. Development 2021; 148:dev.192658. [PMID: 33334861 PMCID: PMC7847271 DOI: 10.1242/dev.192658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/10/2020] [Indexed: 11/20/2022]
Abstract
The paired pharyngeal arch arteries (PAAs) are transient blood vessels connecting the heart with the dorsal aorta during embryogenesis. Although PAA malformations often occur along with pharyngeal pouch defects, the functional interaction between these adjacent tissues remains largely unclear. Here, we report that pharyngeal pouches are essential for PAA progenitor specification in zebrafish embryos. We reveal that the segmentation of pharyngeal pouches coincides spatiotemporally with the emergence of PAA progenitor clusters. These pouches physically associate with pharyngeal mesoderm in discrete regions and provide a niche microenvironment for PAA progenitor commitment by expressing BMP proteins. Specifically, pouch-derived BMP2a and BMP5 are the primary niche cues responsible for activating the BMP/Smad pathway in pharyngeal mesoderm, thereby promoting progenitor specification. In addition, BMP2a and BMP5 play an inductive function in the expression of the cloche gene npas4l in PAA progenitors. cloche mutants exhibit a striking failure to specify PAA progenitors and display ectopic expression of head muscle markers in the pharyngeal mesoderm. Therefore, our results support a crucial role for pharyngeal pouches in establishing a progenitor niche for PAA morphogenesis via BMP2a/5 expression.
Collapse
Affiliation(s)
- Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
18
|
Moreno-Ayala R, Olivares-Chauvet P, Schäfer R, Junker JP. Variability of an Early Developmental Cell Population Underlies Stochastic Laterality Defects. Cell Rep 2021; 34:108606. [PMID: 33440143 PMCID: PMC7809618 DOI: 10.1016/j.celrep.2020.108606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/28/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Embryonic development seemingly proceeds with almost perfect precision. However, it is largely unknown how much underlying microscopic variability is compatible with normal development. Here, we quantify embryo-to-embryo variability in vertebrate development by studying cell number variation in the zebrafish endoderm. We notice that the size of a sub-population of the endoderm, the dorsal forerunner cells (DFCs, which later form the left-right organizer), exhibits significantly more embryo-to-embryo variation than the rest of the endoderm. We find that, with incubation of the embryos at elevated temperature, the frequency of left-right laterality defects is increased drastically in embryos with a low number of DFCs. Furthermore, we observe that these fluctuations have a large stochastic component among fish of the same genetic background. Hence, a stochastic variation in early development leads to a remarkably strong macroscopic phenotype. These fluctuations appear to be associated with maternal effects in the specification of the DFCs.
High embryo-to-embryo variability of dorsal forerunner cell numbers Fluctuations of dorsal forerunner cells have a large stochastic component Embryos with fewer dorsal forerunner cells frequently develop laterality defects Variability of dorsal forerunner cell numbers is associated to maternal effects
Collapse
Affiliation(s)
- Roberto Moreno-Ayala
- Max Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Hannoversche Strasse 28, 10115 Berlin, Germany
| | - Pedro Olivares-Chauvet
- Max Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Hannoversche Strasse 28, 10115 Berlin, Germany
| | - Ronny Schäfer
- Max Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Hannoversche Strasse 28, 10115 Berlin, Germany
| | - Jan Philipp Junker
- Max Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Hannoversche Strasse 28, 10115 Berlin, Germany.
| |
Collapse
|
19
|
Pak B, Schmitt CE, Choi W, Kim JD, Han O, Alsiö J, Jung DW, Williams DR, Coppieters W, Stainier DYR, Jin SW. Analyses of Avascular Mutants Reveal Unique Transcriptomic Signature of Non-conventional Endothelial Cells. Front Cell Dev Biol 2020; 8:589717. [PMID: 33330468 PMCID: PMC7719722 DOI: 10.3389/fcell.2020.589717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells appear to emerge from diverse progenitors. However, to which extent their developmental origin contributes to define their cellular and molecular characteristics remains largely unknown. Here, we report that a subset of endothelial cells that emerge from the tailbud possess unique molecular characteristics that set them apart from stereotypical lateral plate mesoderm (LPM)-derived endothelial cells. Lineage tracing shows that these tailbud-derived endothelial cells arise at mid-somitogenesis stages, and surprisingly do not require Npas4l or Etsrp function, indicating that they have distinct spatiotemporal origins and are regulated by distinct molecular mechanisms. Microarray and single cell RNA-seq analyses reveal that somitogenesis- and neurogenesis-associated transcripts are over-represented in these tailbud-derived endothelial cells, suggesting that they possess a unique transcriptomic signature. Taken together, our results further reveal the diversity of endothelial cells with respect to their developmental origin and molecular properties, and provide compelling evidence that the molecular characteristics of endothelial cells may reflect their distinct developmental history.
Collapse
Affiliation(s)
- Boryeong Pak
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Christopher E. Schmitt
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Yale Cardiovascular Research Center and Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Woosoung Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Jun-Dae Kim
- Yale Cardiovascular Research Center and Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Orjin Han
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Jessica Alsiö
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Da-Woon Jung
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Darren R. Williams
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Wouter Coppieters
- Unit of Animal Genomics, Faculty of Veterinary Medicine, Interdisciplinary Institute of Applied Genomics (GIGA-R), University of Liège (B34), Liège, Belgium
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Suk-Won Jin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Yale Cardiovascular Research Center and Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
20
|
Schumacher JA, Wright ZA, Owen ML, Bredemeier NO, Sumanas S. Integrin α5 and Integrin α4 cooperate to promote endocardial differentiation and heart morphogenesis. Dev Biol 2020; 465:46-57. [PMID: 32628938 DOI: 10.1016/j.ydbio.2020.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 10/23/2022]
Abstract
Endocardium is critically important for proper function of the cardiovascular system. Not only does endocardium connect the heart to blood vasculature, it also plays an important role in heart morphogenesis, valve formation, and ventricular trabeculation. The extracellular protein Fibronectin (Fn1) promotes endocardial differentiation, but the signaling pathways downstream of Fn1 that regulate endocardial development are not understood. Here, we analyzed the role of the Fibronectin receptors Integrin alpha5 (Itga5) and Integrin alpha4 (Itga4) in zebrafish heart development. We show that itga5 mRNA is expressed in both endocardium and myocardium during early stages of heart development. Through analysis of both itga5 single mutants and itga4;itga5 double mutants, we show that loss of both itga5 and itga4 results in enhanced defects in endocardial differentiation and morphogenesis compared to loss of itga5 alone. Loss of both itga5 and itga4 results in cardia bifida and severe myocardial morphology defects. Finally, we find that loss of itga5 and itga4 results in abnormally narrow anterior endodermal sheet morphology. Together, our results support a model in which Itga5 and Itga4 cooperate to promote endocardial differentiation, medial migration of endocardial and myocardial cells, and morphogenesis of anterior endoderm.
Collapse
Affiliation(s)
- Jennifer A Schumacher
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Biological Sciences, Miami University, Hamilton, OH, USA.
| | - Zoë A Wright
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mackenzie L Owen
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nina O Bredemeier
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
21
|
Abstract
The lateral plate mesoderm (LPM) forms the progenitor cells that constitute the heart and cardiovascular system, blood, kidneys, smooth muscle lineage and limb skeleton in the developing vertebrate embryo. Despite this central role in development and evolution, the LPM remains challenging to study and to delineate, owing to its lineage complexity and lack of a concise genetic definition. Here, we outline the processes that govern LPM specification, organization, its cell fates and the inferred evolutionary trajectories of LPM-derived tissues. Finally, we discuss the development of seemingly disparate organ systems that share a common LPM origin. Summary: The lateral plate mesoderm is the origin of several major cell types and organ systems in the vertebrate body plan. How this mesoderm territory emerges and partitions into its downstream fates provides clues about vertebrate development and evolution.
Collapse
Affiliation(s)
- Karin D Prummel
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA.,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Susan Nieuwenhuize
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA.,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA .,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| |
Collapse
|
22
|
Mao A, Zhang M, Liu J, Cao Y, Wang Q. PDGF signaling from pharyngeal pouches promotes arch artery morphogenesis. J Genet Genomics 2019; 46:551-559. [DOI: 10.1016/j.jgg.2019.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/05/2019] [Accepted: 11/07/2019] [Indexed: 01/16/2023]
|
23
|
Hernandez L, Ryckebüsch L, Wang C, Ling R, Yelon D. Tmem2 restricts atrioventricular canal differentiation by regulating degradation of hyaluronic acid. Dev Dyn 2019; 248:1195-1210. [DOI: 10.1002/dvdy.106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/02/2019] [Accepted: 08/04/2019] [Indexed: 11/12/2022] Open
Affiliation(s)
- Lydia Hernandez
- Division of Biological SciencesUniversity of California, San Diego La Jolla California
| | - Lucile Ryckebüsch
- Division of Biological SciencesUniversity of California, San Diego La Jolla California
| | - Carole Wang
- Division of Biological SciencesUniversity of California, San Diego La Jolla California
| | - Rachel Ling
- Division of Biological SciencesUniversity of California, San Diego La Jolla California
| | - Deborah Yelon
- Division of Biological SciencesUniversity of California, San Diego La Jolla California
| |
Collapse
|
24
|
Abstract
Soon after fertilization the zebrafish embryo generates the pool of cells that will give rise to the germline and the three somatic germ layers of the embryo (ectoderm, mesoderm and endoderm). As the basic body plan of the vertebrate embryo emerges, evolutionarily conserved developmental signaling pathways, including Bmp, Nodal, Wnt, and Fgf, direct the nearly totipotent cells of the early embryo to adopt gene expression profiles and patterns of cell behavior specific to their eventual fates. Several decades of molecular genetics research in zebrafish has yielded significant insight into the maternal and zygotic contributions and mechanisms that pattern this vertebrate embryo. This new understanding is the product of advances in genetic manipulations and imaging technologies that have allowed the field to probe the cellular, molecular and biophysical aspects underlying early patterning. The current state of the field indicates that patterning is governed by the integration of key signaling pathways and physical interactions between cells, rather than a patterning system in which distinct pathways are deployed to specify a particular cell fate. This chapter focuses on recent advances in our understanding of the genetic and molecular control of the events that impart cell identity and initiate the patterning of tissues that are prerequisites for or concurrent with movements of gastrulation.
Collapse
Affiliation(s)
- Florence L Marlow
- Icahn School of Medicine Mount Sinai Department of Cell, Developmental and Regenerative Biology, New York, NY, United States.
| |
Collapse
|
25
|
Prummel KD, Hess C, Nieuwenhuize S, Parker HJ, Rogers KW, Kozmikova I, Racioppi C, Brombacher EC, Czarkwiani A, Knapp D, Burger S, Chiavacci E, Shah G, Burger A, Huisken J, Yun MH, Christiaen L, Kozmik Z, Müller P, Bronner M, Krumlauf R, Mosimann C. A conserved regulatory program initiates lateral plate mesoderm emergence across chordates. Nat Commun 2019; 10:3857. [PMID: 31451684 PMCID: PMC6710290 DOI: 10.1038/s41467-019-11561-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/22/2019] [Indexed: 01/06/2023] Open
Abstract
Cardiovascular lineages develop together with kidney, smooth muscle, and limb connective tissue progenitors from the lateral plate mesoderm (LPM). How the LPM initially emerges and how its downstream fates are molecularly interconnected remain unknown. Here, we isolate a pan-LPM enhancer in the zebrafish-specific draculin (drl) gene that provides specific LPM reporter activity from early gastrulation. In toto live imaging and lineage tracing of drl-based reporters captures the dynamic LPM emergence as lineage-restricted mesendoderm field. The drl pan-LPM enhancer responds to the transcription factors EomesoderminA, FoxH1, and MixL1 that combined with Smad activity drive LPM emergence. We uncover specific activity of zebrafish-derived drl reporters in LPM-corresponding territories of several chordates including chicken, axolotl, lamprey, Ciona, and amphioxus, revealing a universal upstream LPM program. Altogether, our work provides a mechanistic framework for LPM emergence as defined progenitor field, possibly representing an ancient mesodermal cell state that predates the primordial vertebrate embryo. Numerous tissues are derived from the lateral plate mesoderm (LPM) but how this is specified is unclear. Here, the authors identify a pan-LPM reporter activity found in the zebrafish draculin (drl) gene that also shows transgenic activity in LPM-corresponding territories of several chordates, including chicken, axolotl, lamprey, Ciona, and amphioxus.
Collapse
Affiliation(s)
- Karin D Prummel
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Christopher Hess
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Susan Nieuwenhuize
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Hugo J Parker
- Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, KS, 66160, USA.,Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Katherine W Rogers
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, 72076, Germany
| | - Iryna Kozmikova
- Institute of Molecular Genetics of the ASCR, Prague, 142 20, Czech Republic
| | - Claudia Racioppi
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, 10003, USA
| | - Eline C Brombacher
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Anna Czarkwiani
- TUD-CRTD Center for Regenerative Therapies Dresden, Dresden, 01307, Germany
| | - Dunja Knapp
- TUD-CRTD Center for Regenerative Therapies Dresden, Dresden, 01307, Germany
| | - Sibylle Burger
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Elena Chiavacci
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Gopi Shah
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Alexa Burger
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland
| | - Jan Huisken
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany.,Morgridge Institute for Research, Madison, WI, 53715, USA
| | - Maximina H Yun
- TUD-CRTD Center for Regenerative Therapies Dresden, Dresden, 01307, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, 10003, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, Prague, 142 20, Czech Republic
| | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, 72076, Germany
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Robb Krumlauf
- Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, KS, 66160, USA.,Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zurich, Zürich, 8057, Switzerland.
| |
Collapse
|
26
|
Li L, Ning G, Yang S, Yan Y, Cao Y, Wang Q. BMP signaling is required for nkx2.3-positive pharyngeal pouch progenitor specification in zebrafish. PLoS Genet 2019; 15:e1007996. [PMID: 30763319 PMCID: PMC6392332 DOI: 10.1371/journal.pgen.1007996] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/27/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
Pharyngeal pouches, a series of outpocketings that bud from the foregut endoderm, are essential to the formation of craniofacial skeleton as well as several important structures like parathyroid and thymus. However, whether pharyngeal pouch progenitors exist in the developing gut tube remains unknown. Here, taking advantage of cell lineage tracing and transgenic ablation technologies, we identified a population of nkx2.3+ pouch progenitors in zebrafish embryos and demonstrated an essential requirement of ectodermal BMP2b for their specification. At early somite stages, nkx2.3+ cells located at lateral region of pharyngeal endoderm give rise to the pouch epithelium except a subpopulation expressing pdgfαa rather than nkx2.3. A small-scale screen of chemical inhibitors reveals that BMP signaling is necessary to specify these progenitors. Loss-of-function analyses show that BMP2b, expressed in the pharyngeal ectoderm, actives Smad effectors in endodermal cells to induce nkx2.3+ progenitors. Collectively, our study provides in vivo evidence for the existence of pouch progenitors and highlights the importance of BMP2b signaling in progenitor specification. Pharyngeal pouches are essential to the formation of craniofacial skeleton as well as several important structures like parathyroid and thymus, but whether their progenitors exist in the developing gut tube remains unknown. Our study provide in vivo evidence that, in the early somite stages, nkx2.3+ cells are present in the lateral pharyngeal endoderm and give rise to the pouch epithelium. We further reveal that ectodermal BMP2b is essential for the activation of Smad effectors in endodermal cells, thereby facilitating pouch progenitor specification. Collectively, our discoveries shed new light on the cellular and molecular mechanisms of pharyngeal pouch development.
Collapse
Affiliation(s)
- Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Shuyan Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yifang Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
27
|
Warga RM, Kane DA. Wilson cell origin for kupffer's vesicle in the zebrafish. Dev Dyn 2018; 247:1057-1069. [DOI: 10.1002/dvdy.24657] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Rachel M. Warga
- Department of Biological Sciences; Western Michigan University; Kalamazoo Michigan
| | - Donald A. Kane
- Department of Biological Sciences; Western Michigan University; Kalamazoo Michigan
| |
Collapse
|
28
|
Hu B, Gao Y, Davies L, Woo S, Topczewski J, Jessen JR, Lin F. Glypican 4 and Mmp14 interact in regulating the migration of anterior endodermal cells by limiting extracellular matrix deposition. Development 2018; 145:dev.163303. [PMID: 30082271 DOI: 10.1242/dev.163303] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/16/2018] [Indexed: 01/30/2023]
Abstract
During embryogenesis, the germ layers, including the endoderm, undergo convergence and extension movements to narrow and elongate the body plan. In zebrafish, the dorsal migration of endodermal cells during gastrulation is controlled by chemokine signaling, but little is known about how they migrate during segmentation. Here, we show that glypican 4 (Gpc4), a member of the heparin sulfate proteoglycan family, is required for efficient migration of anterior endodermal cells during early segmentation, regulating Rac activation to maintain polarized actin-rich lamellipodia. An endoderm transplantation assay showed that Gpc4 regulates endoderm migration in a non-cell-autonomous fashion. Further analyses revealed that the impaired endoderm migration in gpc4 mutants results from increases in the expression and assembly of fibronectin and laminin, major components of the extracellular matrix (ECM). Notably, we found that matrix metalloproteinase 14 (Mmp14a/b) is required for the control of ECM expression during endoderm migration, with Gpc4 acting through Mmp14a/b to limit ECM expression. Our results suggest that Gpc4 is crucial for generating the environment required for efficient migration of endodermal cells, uncovering a novel function of Gpc4 during development.
Collapse
Affiliation(s)
- Bo Hu
- 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
| | - Lauren Davies
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Stephanie Woo
- School of Natural Sciences, Merced, University of California Merced, Merced, CA 95340, USA
| | - Jacek Topczewski
- Northwestern University, Feinberg School of Medicine, Stanley Manne Children's Research Institute, Chicago, IL 60611, USA.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| | - Jason R Jessen
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
29
|
Veil M, Schaechtle MA, Gao M, Kirner V, Buryanova L, Grethen R, Onichtchouk D. Maternal Nanog is required for zebrafish embryo architecture and for cell viability during gastrulation. Development 2018; 145:dev.155366. [PMID: 29180568 DOI: 10.1242/dev.155366] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022]
Abstract
Nanog has been implicated in establishment of pluripotency in mammals and in zygotic genome activation in zebrafish. In this study, we characterize the development of MZnanog (maternal and zygotic null) mutant zebrafish embryos. Without functional Nanog, epiboly is severely affected, embryo axes do not form and massive cell death starts at the end of gastrulation. We show that three independent defects in MZnanog mutants contribute to epiboly failure: yolk microtubule organization required for epiboly is abnormal, maternal mRNA fails to degrade owing to the absence of miR-430, and actin structure of the yolk syncytial layer does not form properly. We further demonstrate that the cell death in MZnanog embryos is cell-autonomous. Nanog is necessary for correct spatial expression of the ventral-specifying genes bmp2b, vox and vent, and the neural transcription factor her3 It is also required for the correctly timed activation of endoderm genes and for the degradation of maternal eomesa mRNA via miR-430. Our findings suggest that maternal Nanog coordinates several gene regulatory networks that shape the embryo during gastrulation.
Collapse
Affiliation(s)
- Marina Veil
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Anna Schaechtle
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Meijiang Gao
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Viola Kirner
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Lenka Buryanova
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Rachel Grethen
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Daria Onichtchouk
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany .,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,Institute of Developmental Biology RAS, 119991 Moscow, Russia
| |
Collapse
|
30
|
Watanabe M, Yasuoka Y, Mawaribuchi S, Kuretani A, Ito M, Kondo M, Ochi H, Ogino H, Fukui A, Taira M, Kinoshita T. Conservatism and variability of gene expression profiles among homeologous transcription factors in Xenopus laevis. Dev Biol 2017; 426:301-324. [DOI: 10.1016/j.ydbio.2016.09.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 07/27/2016] [Accepted: 09/19/2016] [Indexed: 12/11/2022]
|
31
|
Lilly AJ, Lacaud G, Kouskoff V. SOXF transcription factors in cardiovascular development. Semin Cell Dev Biol 2017; 63:50-57. [PMID: 27470491 DOI: 10.1016/j.semcdb.2016.07.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/19/2016] [Accepted: 07/23/2016] [Indexed: 12/24/2022]
Abstract
Cardiovascular development during embryogenesis involves complex changes in gene regulatory networks regulated by a variety of transcription factors. In this review we discuss the various reported roles of the SOXF factors: SOX7, SOX17 and SOX18 in cardiac, vascular and lymphatic development. SOXF factors have pleiotropic roles during these processes, and there is significant redundancy and functional compensation between SOXF family members. Despite this, evidence suggests that there is some specificity in the transcriptional programs they regulate which is necessary to control the differentiation and behaviour of endothelial subpopulations. Furthermore, SOXF factors appear to have an indirect role in regulating cardiac mesoderm specification and differentiation. Understanding how SOXF factors are regulated, as well as their downstream transcriptional target genes, will be important for unravelling their roles in cardiovascular development and related diseases.
Collapse
Affiliation(s)
- Andrew J Lilly
- Cancer Research UK, Stem Cell Hematopoiesis, The University of Manchester, Wilmslow road, M20 4BX, UK
| | - Georges Lacaud
- Cancer Research UK, Stem Cell Biology group Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK.
| | - Valerie Kouskoff
- Cancer Research UK, Stem Cell Hematopoiesis, The University of Manchester, Wilmslow road, M20 4BX, UK.
| |
Collapse
|
32
|
Villasenor A, Stainier DYR. On the development of the hepatopancreatic ductal system. Semin Cell Dev Biol 2017; 66:69-80. [PMID: 28214561 DOI: 10.1016/j.semcdb.2017.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
The hepatopancreatic ductal system is the collection of ducts that connect the liver and pancreas to the digestive tract. The formation of this system is necessary for the transport of exocrine secretions, for the correct assembly of the pancreatobiliary ductal system, and for the overall function of the digestive system. Studies on endoderm organ formation have significantly advanced our understanding of the molecular mechanisms that govern organ induction, organ specification and morphogenesis of the major foregut-derived organs. However, little is known about the mechanisms that control the development of the hepatopancreatic ductal system. Here, we provide a description of the different components of the system, summarize its development from the endoderm to a complex system of tubes, list the pathologies produced by anomalies in its development, as well as the molecules and signaling pathways that are known to be involved in its formation. Finally, we discuss its proposed potential as a multipotent cell reservoir and the unresolved questions in the field.
Collapse
Affiliation(s)
- Alethia Villasenor
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
33
|
Bloomekatz J, Singh R, Prall OW, Dunn AC, Vaughan M, Loo CS, Harvey RP, Yelon D. Platelet-derived growth factor (PDGF) signaling directs cardiomyocyte movement toward the midline during heart tube assembly. eLife 2017; 6:21172. [PMID: 28098558 PMCID: PMC5298878 DOI: 10.7554/elife.21172] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Communication between neighboring tissues plays a central role in guiding organ morphogenesis. During heart tube assembly, interactions with the adjacent endoderm control the medial movement of cardiomyocytes, a process referred to as cardiac fusion. However, the molecular underpinnings of this endodermal-myocardial relationship remain unclear. Here, we show an essential role for platelet-derived growth factor receptor alpha (Pdgfra) in directing cardiac fusion. Mutation of pdgfra disrupts heart tube assembly in both zebrafish and mouse. Timelapse analysis of individual cardiomyocyte trajectories reveals misdirected cells in zebrafish pdgfra mutants, suggesting that PDGF signaling steers cardiomyocytes toward the midline during cardiac fusion. Intriguingly, the ligand pdgfaa is expressed in the endoderm medial to the pdgfra-expressing myocardial precursors. Ectopic expression of pdgfaa interferes with cardiac fusion, consistent with an instructive role for PDGF signaling. Together, these data uncover a novel mechanism through which endodermal-myocardial communication can guide the cell movements that initiate cardiac morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.21172.001 In the growing embryo, the heart initially develops in the form of a simple tube. Its outer layer is made up of muscular cells, called myocardial cells, that pump blood through the tube. Before the heart tube develops, two groups of myocardial cells exist – one on each side of the embryo. To assemble the heart, these two populations of cells must move as a group to the middle of the embryo, where they meet and merge through a process called cardiac fusion. This movement of myocardial cells toward the middle of the embryo depends upon interactions with a neighboring tissue called the endoderm. How the endoderm directs the movement of the myocardial cells was not well understood. The PDGF signaling pathway guides the movement of several different types of cells in the body, but it had not been previously linked to the early stages of heart tube assembly. In this pathway, a molecule called platelet-derived growth factor (PDGF) binds to PDGF receptors that sit on the surface of cells. Using microscopy and genetic analysis to study zebrafish and mouse embryos, Bloomekatz et al. now show that embryos that carry mutations in a gene that encodes a PDGF receptor suffer from defects in heart tube assembly. Further examination of the mutant zebrafish embryos revealed that the myocardial cells were not properly directed toward the middle of the embryo. In fact, many of these cells appeared to move away from the midline. Bloomekatz et al. also observed that, in normal embryos, the endoderm cells that lie adjacent to the myocardial cells produce PDGF. Therefore, it appears that PDGF produced by the endoderm could interact with PDGF receptors on the myocardial cells to direct these cells toward the middle of the embryo. The next step will be to figure out how this signaling influences the machinery inside the myocardial cells that controls their movement. Ultimately, this knowledge could lead to new ways to identify and treat congenital heart diseases. DOI:http://dx.doi.org/10.7554/eLife.21172.002
Collapse
Affiliation(s)
- Joshua Bloomekatz
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Reena Singh
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, Australia
| | - Owen Wj Prall
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ariel C Dunn
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Megan Vaughan
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Chin-San Loo
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, Australia
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|
34
|
Hozumi S, Aoki S, Kikuchi Y. Nuclear movement regulated by non-Smad Nodal signaling via JNK is associated with Smad signaling during zebrafish endoderm specification. Development 2017; 144:4015-4025. [DOI: 10.1242/dev.151746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/14/2017] [Indexed: 02/01/2023]
Abstract
Although asymmetric nuclear positioning is observed during animal development, the regulation and significance of this nuclear positioning in cell differentiation remains poorly understood. Using zebrafish blastulae, we provide evidence that nuclear movement toward the yolk syncytial layer, which comprises extraembryonic tissue, occurs in the first endoderm specified cells during endoderm specification. Nodal signaling is essential for nuclear movement, whereas nuclear envelope proteins are involved in the movement through the microtubule formation. The positioning of the microtubule organizing center, which is proposed to be critical for nuclear movement, is regulated by Nodal signaling and nuclear envelope proteins. The non-Smad JNK signaling pathway, which is downstream of Nodal signaling, regulates nuclear movement independent of the Smad pathway, and this nuclear movement is associated with Smad signal transduction toward the nucleus. Our study provides insights into the function of nuclear movement in Smad signaling toward the nucleus, and could be applied to the control of Transforming Growth Factor-β signaling.
Collapse
Affiliation(s)
- Shunya Hozumi
- Department of Biological Science, Graduate School of Science, Hiroshima University, Kagamiyama 1-3-1, Higashi-Hiroshima, Hiroshima, 739-8526 Japan
| | - Shun Aoki
- Department of Biological Science, Graduate School of Science, Hiroshima University, Kagamiyama 1-3-1, Higashi-Hiroshima, Hiroshima, 739-8526 Japan
| | - Yutaka Kikuchi
- Department of Biological Science, Graduate School of Science, Hiroshima University, Kagamiyama 1-3-1, Higashi-Hiroshima, Hiroshima, 739-8526 Japan
| |
Collapse
|
35
|
Reade A, Motta-Mena LB, Gardner KH, Stainier DY, Weiner OD, Woo S. TAEL: a zebrafish-optimized optogenetic gene expression system with fine spatial and temporal control. Development 2016; 144:345-355. [PMID: 27993986 DOI: 10.1242/dev.139238] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 11/28/2016] [Indexed: 01/27/2023]
Abstract
Here, we describe an optogenetic gene expression system optimized for use in zebrafish. This system overcomes the limitations of current inducible expression systems by enabling robust spatial and temporal regulation of gene expression in living organisms. Because existing optogenetic systems show toxicity in zebrafish, we re-engineered the blue-light-activated EL222 system for minimal toxicity while exhibiting a large range of induction, fine spatial precision and rapid kinetics. We validate several strategies to spatially restrict illumination and thus gene induction with our new TAEL (TA4-EL222) system. As a functional example, we show that TAEL is able to induce ectopic endodermal cells in the presumptive ectoderm via targeted sox32 induction. We also demonstrate that TAEL can be used to resolve multiple roles of Nodal signaling at different stages of embryonic development. Finally, we show how inducible gene editing can be achieved by combining the TAEL and CRISPR/Cas9 systems. This toolkit should be a broadly useful resource for the fish community.
Collapse
Affiliation(s)
- Anna Reade
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Laura B Motta-Mena
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, NY 10031, USA
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, NY 10031, USA.,Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031, USA.,Biochemistry, Chemistry and Biology PhD Programs, Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Didier Y Stainier
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.,Max Planck Institute for Heart and Lung Research, Dept. of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Orion D Weiner
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephanie Woo
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
36
|
Perez-Camps M, Tian J, Chng SC, Sem KP, Sudhaharan T, Teh C, Wachsmuth M, Korzh V, Ahmed S, Reversade B. Quantitative imaging reveals real-time Pou5f3-Nanog complexes driving dorsoventral mesendoderm patterning in zebrafish. eLife 2016; 5. [PMID: 27684073 PMCID: PMC5042653 DOI: 10.7554/elife.11475] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 08/04/2016] [Indexed: 12/11/2022] Open
Abstract
Formation of the three embryonic germ layers is a fundamental developmental process that initiates differentiation. How the zebrafish pluripotency factor Pou5f3 (homologous to mammalian Oct4) drives lineage commitment is unclear. Here, we introduce fluorescence lifetime imaging microscopy and fluorescence correlation spectroscopy to assess the formation of Pou5f3 complexes with other transcription factors in real-time in gastrulating zebrafish embryos. We show, at single-cell resolution in vivo, that Pou5f3 complexes with Nanog to pattern mesendoderm differentiation at the blastula stage. Later, during gastrulation, Sox32 restricts Pou5f3–Nanog complexes to the ventrolateral mesendoderm by binding Pou5f3 or Nanog in prospective dorsal endoderm. In the ventrolateral endoderm, the Elabela / Aplnr pathway limits Sox32 levels, allowing the formation of Pou5f3–Nanog complexes and the activation of downstream BMP signaling. This quantitative model shows that a balance in the spatiotemporal distribution of Pou5f3–Nanog complexes, modulated by Sox32, regulates mesendoderm specification along the dorsoventral axis. DOI:http://dx.doi.org/10.7554/eLife.11475.001 As an animal embryo develops, cells divide and establish three distinct layers called the ectoderm, mesoderm and endoderm. Proteins called transcription factors control this process by regulating the activity of particular genes. Two or more transcription factors may interact to modulate each other’s activity. Zebrafish embryos provide an ideal model system for monitoring how these embryonic layers form and the interactions between transcription factors in real-time because they are transparent and develop outside their parents. Pou5f3 and Nanog are two key transcription factors involved in this process in zebrafish. However, it is not clear how Pou5f3 and Nanog instruct cells to become ectoderm, mesoderm or endoderm. Perez Camps et al. used imaging techniques to study Pou5f3 and Nanog. The experiments show that Pou5f3 and Nanog bind together to form complexes that instruct cells to form the temporary layer that later gives rise to both the mesoderm and endoderm. The cells in which there are less Pou5f3 and Nanog complexes form the ectoderm layer. To develop the body shape of adult zebrafish, the embryos need to give individual cells information about their location in the body. For example, a signal protein called bone morphogenetic protein (BMP) accumulates on the side of the embryo that will become the underside of the fish. Perez Camps et al. show that once the endoderm, mesoderm and ectoderm have formed, Pou5f3–Nanog complexes regulate BMP signalling to specify the underside of the fish. Meanwhile, in the endoderm on the opposite side, another transcription factor called Sox32 binds to individual Pou5f3 and Nanog proteins. This prevents Pou5f3 and Nanog from forming complexes and determines which side of the embryo will make the topside of the fish. A future challenge is to explore other transcription factors that may prevent Pou5f1 and Nanog from binding in the mesoderm and ectoderm of the topside of the fish. DOI:http://dx.doi.org/10.7554/eLife.11475.002
Collapse
Affiliation(s)
| | - Jing Tian
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Serene C Chng
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Kai Pin Sem
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | | | - Cathleen Teh
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Malte Wachsmuth
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Sohail Ahmed
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Bruno Reversade
- Institute of Medical Biology, A*STAR, Singapore, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| |
Collapse
|
37
|
Liu Z, Ning G, Xu R, Cao Y, Meng A, Wang Q. Fscn1 is required for the trafficking of TGF-β family type I receptors during endoderm formation. Nat Commun 2016; 7:12603. [PMID: 27545838 PMCID: PMC4996939 DOI: 10.1038/ncomms12603] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/15/2016] [Indexed: 01/01/2023] Open
Abstract
Microtubules function in TGF-β signalling by facilitating the cytoplasmic trafficking of internalized receptors and the nucleocytoplasmic shuttling of Smads. However, nothing is known about whether actin filaments are required for these processes. Here we report that zebrafish actin-bundling protein fscn1a is highly expressed in mesendodermal precursors and its expression is directly regulated by the TGF-β superfamily member Nodal. Knockdown or knockout of fscn1a leads to a reduction of Nodal signal transduction and endoderm formation in zebrafish embryos. Fscn1 specifically interacts with TGF-β family type I receptors, and its depletion disrupts the association between receptors and actin filaments and sequesters the internalized receptors into clathrin-coated vesicles. Therefore, Fscn1 acts as a molecular linker between TGF-β family type I receptors and the actin filaments to promote the trafficking of internalized receptors from clathrin-coated vesicles to early endosomes during zebrafish endoderm formation. It is unclear how the cytoskeleton acts to assist in TGF-β signalling downstream of the receptor. Here, in zebrafish, the authors show that the actin-bundling protein FSCN1 interacts with TGF-β type I receptors ALK 4 and 5, enabling actin filament mediated vesicle trafficking and endoderm formation.
Collapse
Affiliation(s)
- Zhaoting Liu
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ranran Xu
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Anming Meng
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
38
|
The endoderm indirectly influences morphogenetic movements of the zebrafish head kidney through the posterior cardinal vein and VegfC. Sci Rep 2016; 6:30677. [PMID: 27477767 PMCID: PMC4967926 DOI: 10.1038/srep30677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/07/2016] [Indexed: 02/01/2023] Open
Abstract
Integration of blood vessels and organ primordia determines organ shape and function. The head kidney in the zebrafish interacts with the dorsal aorta (DA) and the posterior cardinal vein (PCV) to achieve glomerular filtration and definitive hematopoiesis, respectively. How the head kidney co-develops with both the axial artery and vein remains unclear. We found that in endodermless sox32-deficient embryos, the head kidney associated with the PCV but not the DA. Disrupted convergent migration of the PCV and the head kidney in sox32-deficient embryos was rescued in a highly coordinated fashion through the restoration of endodermal cells. Moreover, grafted endodermal cells abutted the host PCV endothelium in the transplantation assay. Interestingly, the severely-disrupted head kidney convergence in the sox32-deficient embryo was suppressed by both the cloche mutation and the knockdown of endothelial genes, indicating that an interaction between the endoderm and the PCV restricts the migration of the head kidney. Furthermore, knockdown of either vegfC or its receptor vegfr3 suppressed the head kidney convergence defect in endodermless embryos and perturbed the head kidney-PCV association in wild-type embryos. Our findings thus underscore a role for PCV and VegfC in patterning the head kidney prior to organ assembly and function.
Collapse
|
39
|
Haack T, Abdelilah-Seyfried S. The force within: endocardial development, mechanotransduction and signalling during cardiac morphogenesis. Development 2016; 143:373-86. [PMID: 26839341 DOI: 10.1242/dev.131425] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.
Collapse
Affiliation(s)
- Timm Haack
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, Potsdam D-14476, Germany
| |
Collapse
|
40
|
Abstract
The endoderm is the innermost embryonic germ layer, and in zebrafish, it gives rise to the lining of the gut, the gills, liver, pancreas, gallbladder, and derivatives of the pharyngeal pouch. These organs form the gastrointestinal tract and are involved with the absorption, delivery, and metabolism of nutrients. The liver has a central role in regulating these processes because it controls carbohydrate and lipid metabolism, protein synthesis, and breakdown of endogenous and xenobiotic products. Liver dysfunction frequently leads to significant morbidity and mortality; however, in most settings of organ injury, the liver exhibits remarkable regenerative capacity. In this chapter, we review the principal mechanisms of endoderm and liver formation and provide protocols to assess liver formation and liver regeneration.
Collapse
|
41
|
Xie H, Ye D, Sepich D, Lin F. S1pr2/Gα13 signaling regulates the migration of endocardial precursors by controlling endoderm convergence. Dev Biol 2016; 414:228-43. [PMID: 27158029 DOI: 10.1016/j.ydbio.2016.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/09/2016] [Accepted: 04/26/2016] [Indexed: 10/21/2022]
Abstract
Formation of the heart tube requires synchronized migration of endocardial and myocardial precursors. Our previous studies indicated that in S1pr2/Gα13-deficient embryos, impaired endoderm convergence disrupted the medial migration of myocardial precursors, resulting in the formation of two myocardial populations. Here we show that endoderm convergence also regulates endocardial migration. In embryos defective for S1pr2/Gα13 signaling, endocardial precursors failed to migrate towards the midline, and the presumptive endocardium surrounded the bilaterally-located myocardial cells rather than being encompassed by them. In vivo imaging of control embryos revealed that, like their myocardial counterparts, endocardial precursors migrated with the converging endoderm, though from a more anterior point, then moved from the dorsal to the ventral side of the endoderm (subduction), and finally migrated posteriorly towards myocardial precursors, ultimately forming the inner layer of the heart tube. In embryos defective for endoderm convergence due to an S1pr2/Gα13 deficiency, both the medial migration and the subduction of endocardial precursors were impaired, and their posterior migration towards the myocardial precursors was premature. This placed them medial to the myocardial populations, physically blocking the medial migration of the myocardial precursors. Furthermore, contact between the endocardial and myocardial precursor populations disrupted the epithelial architecture of the myocardial precursors, and thus their medial migration; in embryos depleted of endocardial cells, the myocardial migration defect was partially rescued. Our data indicate that endoderm convergence regulates the medial migration of endocardial precursors, and that premature association of the endocardial and myocardial populations contributes to myocardial migration defects observed in S1pr2/Gα13-deficient embryos. The demonstration that endoderm convergence regulates the synchronized migration of endocardial and myocardial precursors reveals a new role of the endoderm in heart development.
Collapse
Affiliation(s)
- Huaping Xie
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 1-400 Bowen Science Building, 51 N Road, Iowa City, IA 52242-1109, USA
| | - Ding Ye
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 1-400 Bowen Science Building, 51 N Road, Iowa City, IA 52242-1109, USA; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Diane Sepich
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 1-400 Bowen Science Building, 51 N Road, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
42
|
Lovely CB, Swartz ME, McCarthy N, Norrie JL, Eberhart JK. Bmp signaling mediates endoderm pouch morphogenesis by regulating Fgf signaling in zebrafish. Development 2016; 143:2000-11. [PMID: 27122171 DOI: 10.1242/dev.129379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 04/12/2016] [Indexed: 02/03/2023]
Abstract
The endodermal pouches are a series of reiterated structures that segment the pharyngeal arches and help pattern the vertebrate face. Multiple pathways regulate the complex process of endodermal development, including the Bone morphogenetic protein (Bmp) pathway. However, the role of Bmp signaling in pouch morphogenesis is poorly understood. Using genetic and chemical inhibitor approaches, we show that pouch morphogenesis requires Bmp signaling from 10-18 h post-fertilization, immediately following gastrulation. Blocking Bmp signaling during this window results in morphological defects to the pouches and craniofacial skeleton. Using genetic chimeras we show that Bmp signals directly to the endoderm for proper morphogenesis. Time-lapse imaging and analysis of reporter transgenics show that Bmp signaling is necessary for pouch outpocketing via the Fibroblast growth factor (Fgf) pathway. Double loss-of-function analyses demonstrate that Bmp and Fgf signaling interact synergistically in craniofacial development. Collectively, our analyses shed light on the tissue and signaling interactions that regulate development of the vertebrate face.
Collapse
Affiliation(s)
- C Ben Lovely
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Mary E Swartz
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Neil McCarthy
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
43
|
Deshwar AR, Chng SC, Ho L, Reversade B, Scott IC. The Apelin receptor enhances Nodal/TGFβ signaling to ensure proper cardiac development. eLife 2016; 5. [PMID: 27077952 PMCID: PMC4859801 DOI: 10.7554/elife.13758] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 04/11/2016] [Indexed: 01/07/2023] Open
Abstract
The Apelin receptor (Aplnr) is essential for heart development, controlling the early migration of cardiac progenitors. Here we demonstrate that in zebrafish Aplnr modulates Nodal/TGFβ signaling, a key pathway essential for mesendoderm induction and migration. Loss of Aplnr function leads to a reduction in Nodal target gene expression whereas activation of Aplnr by a non-peptide agonist increases the expression of these same targets. Furthermore, loss of Aplnr results in a delay in the expression of the cardiogenic transcription factors mespaa/ab. Elevating Nodal levels in aplnra/b morphant and double mutant embryos is sufficient to rescue cardiac differentiation defects. We demonstrate that loss of Aplnr attenuates the activity of a point source of Nodal ligands Squint and Cyclops in a non-cell autonomous manner. Our results favour a model in which Aplnr is required to fine-tune Nodal output, acting as a specific rheostat for the Nodal/TGFβ pathway during the earliest stages of cardiogenesis. DOI:http://dx.doi.org/10.7554/eLife.13758.001 In one of the first events that happens as an embryo develops, cells become the different stem cell populations that form the body’s organs. So what makes a cell become one stem cell type rather than another? In the case of the heart, the first important event is the activity of a signaling pathway called the Nodal/TGFβ pathway. Nodal signaling can drive cells to become many different stem cell types depending on its level of activity. Many different levels of regulation fine-tune Nodal signaling to produce these activity thresholds. Zebrafish that have a mutation in the gene that encodes a protein called the Apelin receptor have no heart. The loss of this receptor interferes with how heart stem cells (called cardiac progenitors) are made and how they move to where heart development occurs. Deshwar et al. have now studied mutant zebrafish in order to investigate how the Apelin receptor influences early heart development. This revealed that Nodal signaling levels are slightly lower in the mutant zebrafish embryos than in normal fish at the time when Nodal activity induces cardiac progenitors to form. When Nodal activity is experimentally boosted in zebrafish that lack the Apelin receptor, they become able to develop hearts. Deshwar et al. also found that the Apelin receptor does not work in cells that produce or receive Nodal signals. This suggests that the Apelin receptor modulates Nodal signaling levels by acting in cells that lie between the cells that release Nodal signals and the cardiac progenitors. An important question for future work to address is how this modulation works. As Nodal is a key determinant of many cell types in developing embryos, learning how Apelin receptors regulate its activity could help researchers to derive specific cell types from cultured stem cells for use in regenerative medicine. DOI:http://dx.doi.org/10.7554/eLife.13758.002
Collapse
Affiliation(s)
- Ashish R Deshwar
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Serene C Chng
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Lena Ho
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Bruno Reversade
- Institute of Medical Biology, A*STAR, Singapore, Singapore.,Institute of Molecular and Cellular Biology, A*STAR, Singapore, Singapore.,Department of Paediatrics, School of Medicine, National University of Singapore, , Singapore
| | - Ian C Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Canada
| |
Collapse
|
44
|
Zhu P, Xu X, Lin X. Both ciliary and non-ciliary functions of Foxj1a confer Wnt/β-catenin signaling in zebrafish left-right patterning. Biol Open 2015; 4:1376-86. [PMID: 26432885 PMCID: PMC4728341 DOI: 10.1242/bio.012088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Wnt/β-catenin pathway is implicated in left-right (LR) axis determination; however, the underlying mechanism remains elusive. Prompted by our recent discovery that Wnt signaling regulates ciliogenesis in the zebrafish Kupffer's vesicle (KV) via Foxj1a, a ciliogenic transcription factor, we decided to elucidate functions of Foxj1a in Wnt-regulated LR pattern formation. We showed that targeted injection of wnt8a mRNA into a single cell at the 128-cell stage is sufficient to induce ectopic foxj1a expression and ectopic cilia. By interrogating the transcription circuit of foxj1a regulation, we found that both Lef1 and Tcf7 bind to a consensus element in the foxj1a promoter region. Depletion of Lef1 and Tcf7 inhibits foxj1a transcription in the dorsal forerunner cells, downregulates cilia length and number in KV, and randomizes LR asymmetry. Targeted overexpression of a constitutively active form of Lef1 also induced an ectopic protrusion that contains ectopic transcripts for sox17, foxj1a, and charon, and ectopic monocilia. Further genetic studies using this ectopic expression platform revealed two distinct functions of Foxj1a; mediating Wnt-governed monocilia length elongation as well as charon transcription. The novel Foxj1a-charon regulation is conserved in KV, and importantly, it is independent of the canonical role of Foxj1a in the biosynthesis of motile cilia. Together with the known function of motile cilia movement in generating asymmetric expression of charon, our data put forward a hypothesis that Foxj1a confers both ciliary and non-ciliary functions of Wnt signaling, which converge on charon to regulate LR pattern formation. Summary: Using a targeted overexpression platform, we showed that Wnt activation induces ectopic foxj1a expression and ectopic cilia formation, and revealed two distinct roles of Foxj1a in conferring Wnt-governed left-right patterning.
Collapse
Affiliation(s)
- Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
45
|
Nicenboim J, Malkinson G, Lupo T, Asaf L, Sela Y, Mayseless O, Gibbs-Bar L, Senderovich N, Hashimshony T, Shin M, Jerafi-Vider A, Avraham-Davidi I, Krupalnik V, Hofi R, Almog G, Astin JW, Golani O, Ben-Dor S, Crosier PS, Herzog W, Lawson ND, Hanna JH, Yanai I, Yaniv K. Lymphatic vessels arise from specialized angioblasts within a venous niche. Nature 2015; 522:56-61. [DOI: 10.1038/nature14425] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/26/2015] [Indexed: 01/02/2023]
|
46
|
Jessen JR. Recent advances in the study of zebrafish extracellular matrix proteins. Dev Biol 2015; 401:110-21. [DOI: 10.1016/j.ydbio.2014.12.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
|
47
|
Liu Y, Kaneda R, Leja TW, Subkhankulova T, Tolmachov O, Minchiotti G, Schwartz RJ, Barahona M, Schneider MD. Hhex and Cer1 mediate the Sox17 pathway for cardiac mesoderm formation in embryonic stem cells. Stem Cells 2015; 32:1515-26. [PMID: 24585688 PMCID: PMC4260090 DOI: 10.1002/stem.1695] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/28/2014] [Accepted: 02/11/2014] [Indexed: 12/11/2022]
Abstract
Cardiac muscle differentiation in vivo is guided by sequential growth factor signals, including endoderm-derived diffusible factors, impinging on cardiogenic genes in the developing mesoderm. Previously, by RNA interference in AB2.2 mouse embryonic stem cells (mESCs), we identified the endodermal transcription factor Sox17 as essential for Mesp1 induction in primitive mesoderm and subsequent cardiac muscle differentiation. However, downstream effectors of Sox17 remained to be proven functionally. In this study, we used genome-wide profiling of Sox17-dependent genes in AB2.2 cells, RNA interference, chromatin immunoprecipitation, and luciferase reporter genes to dissect this pathway. Sox17 was required not only for Hhex (a second endodermal transcription factor) but also for Cer1, a growth factor inhibitor from endoderm that, like Hhex, controls mesoderm patterning in Xenopus toward a cardiac fate. Suppressing Hhex or Cer1 blocked cardiac myogenesis, although at a later stage than induction of Mesp1/2. Hhex was required but not sufficient for Cer1 expression. Over-expression of Sox17 induced endogenous Cer1 and sequence-specific transcription of a Cer1 reporter gene. Forced expression of Cer1 was sufficient to rescue cardiac differentiation in Hhex-deficient cells. Thus, Hhex and Cer1 are indispensable components of the Sox17 pathway for cardiopoiesis in mESCs, acting at a stage downstream from Mesp1/2.
Collapse
Affiliation(s)
- Yu Liu
- Center for Cardiovascular Development, Baylor College of Medicine, Houston, Texas, USA; Institute for Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Verleyen D, Luyten FP, Tylzanowski P. Orphan G-protein coupled receptor 22 (Gpr22) regulates cilia length and structure in the zebrafish Kupffer's vesicle. PLoS One 2014; 9:e110484. [PMID: 25335082 PMCID: PMC4204907 DOI: 10.1371/journal.pone.0110484] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/16/2014] [Indexed: 02/06/2023] Open
Abstract
GPR22 is an orphan G protein-coupled receptor (GPCR). Since the ligand of the receptor is currently unknown, its biological function has not been investigated in depth. Many GPCRs and their intracellular effectors are targeted to cilia. Cilia are highly conserved eukaryotic microtubule-based organelles that protrude from the membrane of most mammalian cells. They are involved in a large variety of physiological processes and diseases. However, the details of the downstream pathways and mechanisms that maintain cilia length and structure are poorly understood. We show that morpholino knock down or overexpression of gpr22 led to defective left-right (LR) axis formation in the zebrafish embryo. Specifically, defective LR patterning included randomization of the left-specific lateral plate mesodermal genes (LPM) (lefty1, lefty2, southpaw and pitx2a), resulting in randomized cardiac looping. Furthermore, gpr22 inactivation in the Kupffer’s vesicle (KV) alone was still able to generate the phenotype, indicating that Gpr22 mainly regulates LR asymmetry through the KV. Analysis of the KV cilia by immunofluorescence and transmission electron microscopy (TEM), revealed that gpr22 knock down or overexpression resulted in changes of cilia length and structure. Further, we found that Gpr22 does not act upstream of the two cilia master regulators, Foxj1a and Rfx2. To conclude, our study characterized a novel player in the field of ciliogenesis.
Collapse
Affiliation(s)
- Daphne Verleyen
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, University of Leuven, Leuven, Belgium
| | - Frank P. Luyten
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, University of Leuven, Leuven, Belgium
| | - Przemko Tylzanowski
- Department of Development and Regeneration, Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Centre, University of Leuven, Leuven, Belgium
- Department of Biochemistry and Molecular Biology, Medical University, Lublin, Poland
- * E-mail:
| |
Collapse
|
49
|
Terashima AV, Mudumana SP, Drummond IA. Odd skipped related 1 is a negative feedback regulator of nodal-induced endoderm development. Dev Dyn 2014; 243:1571-80. [PMID: 25233796 DOI: 10.1002/dvdy.24191] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 09/03/2014] [Accepted: 09/10/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Early embryo patterning is orchestrated by tightly regulated morphogen gradients. The Nodal morphogen patterns the mesendoderm, giving rise to all endoderm and head and trunk mesoderm. High Nodal concentrations favor endoderm differentiation while lower promote mesoderm differentiation. Nodal signaling is controlled by both positive and negative feedback regulation to ensure robust developmental patterning. RESULTS Here we identify odd skipped related 1 (osr1), a zinc finger transcription factor, as a new element in Nodal feedback regulation affecting endoderm development. We show that osr1 expression in zebrafish germ ring mesendoderm requires Nodal signaling; osr1 expression was lost in embryos lacking Nodal signaling. Conversely, osr1 expression was ectopically induced by the activation of Nodal signaling. Furthermore we demonstrate that osr1 responds directly to Nodal signaling. Additionally, osr1 knockdown generated excess endoderm cells marked by sox32 expression while expression of osr1 mRNA was not affected in sox32-deficient embryos. CONCLUSIONS Our findings identify osr1 as a Nodal-induced, negative feedback regulator of Nodal signaling that acts at the earliest stages of endoderm differentiation to limit the number of endoderm progenitors. As such, we propose that osr1 represents a novel network motif controlling the output of Nodal signaling to regulate mesendoderm patterning.
Collapse
|
50
|
Nelson AC, Cutty SJ, Niini M, Stemple DL, Flicek P, Houart C, Bruce AEE, Wardle FC. Global identification of Smad2 and Eomesodermin targets in zebrafish identifies a conserved transcriptional network in mesendoderm and a novel role for Eomesodermin in repression of ectodermal gene expression. BMC Biol 2014; 12:81. [PMID: 25277163 PMCID: PMC4206766 DOI: 10.1186/s12915-014-0081-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Indexed: 12/27/2022] Open
Abstract
Background Nodal signalling is an absolute requirement for normal mesoderm and endoderm formation in vertebrate embryos, yet the transcriptional networks acting directly downstream of Nodal and the extent to which they are conserved is largely unexplored, particularly in vivo. Eomesodermin also plays a role in patterning mesoderm and endoderm in vertebrates, but its mechanisms of action and how it interacts with the Nodal signalling pathway are still unclear. Results Using a combination of expression analysis and chromatin immunoprecipitation with deep sequencing (ChIP-seq) we identify direct targets of Smad2, the effector of Nodal signalling in blastula stage zebrafish embryos, including many novel target genes. Through comparison of these data with published ChIP-seq data in human, mouse and Xenopus we show that the transcriptional network driven by Smad2 in mesoderm and endoderm is conserved in these vertebrate species. We also show that Smad2 and zebrafish Eomesodermin a (Eomesa) bind common genomic regions proximal to genes involved in mesoderm and endoderm formation, suggesting Eomesa forms a general component of the Smad2 signalling complex in zebrafish. Combinatorial perturbation of Eomesa and Smad2-interacting factor Foxh1 results in loss of both mesoderm and endoderm markers, confirming the role of Eomesa in endoderm formation and its functional interaction with Foxh1 for correct Nodal signalling. Finally, we uncover a novel role for Eomesa in repressing ectodermal genes in the early blastula. Conclusions Our data demonstrate that evolutionarily conserved developmental functions of Nodal signalling occur through maintenance of the transcriptional network directed by Smad2. This network is modulated by Eomesa in zebrafish which acts to promote mesoderm and endoderm formation in combination with Nodal signalling, whilst Eomesa also opposes ectoderm gene expression. Eomesa, therefore, regulates the formation of all three germ layers in the early zebrafish embryo. Electronic supplementary material The online version of this article (doi:10.1186/s12915-014-0081-5) contains supplementary material, which is available to authorized users.
Collapse
|