1
|
Sampilo NF, Song JL. microRNA-1 regulates sea urchin skeletogenesis by directly targeting skeletogenic genes and modulating components of signaling pathways. Dev Biol 2024; 508:123-137. [PMID: 38290645 PMCID: PMC10985635 DOI: 10.1016/j.ydbio.2024.01.010] [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: 05/08/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
microRNAs are evolutionarily conserved non-coding RNAs that direct post-transcriptional regulation of target transcripts. In vertebrates, microRNA-1 (miR-1) is expressed in muscle and has been found to play critical regulatory roles in vertebrate angiogenesis, a process that has been proposed to be analogous to sea urchin skeletogenesis. Results indicate that both miR-1 inhibitor and miR-1 mimic-injected larvae have significantly less F-actin enriched circumpharyngeal muscle fibers and fewer gut contractions. In addition, miR-1 regulates the positioning of skeletogenic primary mesenchyme cells (PMCs) and skeletogenesis of the sea urchin embryo. Interestingly, the gain-of-function of miR-1 leads to more severe PMC patterning and skeletal branching defects than its loss-of-function. The results suggest that miR-1 directly suppresses Ets1/2, Tbr, and VegfR7 of the skeletogenic gene regulatory network, and Nodal, and Wnt1 signaling components. This study identifies potential targets of miR-1 that impacts skeletogenesis and muscle formation and contributes to a deeper understanding of miR-1's function during development.
Collapse
Affiliation(s)
- Nina Faye Sampilo
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Jia L Song
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
2
|
Yamakawa S, Yamazaki A, Morino Y, Wada H. Early expression onset of tissue-specific effector genes during the specification process in sea urchin embryos. EvoDevo 2023; 14:7. [PMID: 37101206 PMCID: PMC10131483 DOI: 10.1186/s13227-023-00210-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/01/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND In the course of animal developmental processes, various tissues are differentiated through complex interactions within the gene regulatory network. As a general concept, differentiation has been considered to be the endpoint of specification processes. Previous works followed this view and provided a genetic control scheme of differentiation in sea urchin embryos: early specification genes generate distinct regulatory territories in an embryo to express a small set of differentiation driver genes; these genes eventually stimulate the expression of tissue-specific effector genes, which provide biological identity to differentiated cells, in each region. However, some tissue-specific effector genes begin to be expressed in parallel with the expression onset of early specification genes, raising questions about the simplistic regulatory scheme of tissue-specific effector gene expression and the current concept of differentiation itself. RESULTS Here, we examined the dynamics of effector gene expression patterns during sea urchin embryogenesis. Our transcriptome-based analysis indicated that many tissue-specific effector genes begin to be expressed and accumulated along with the advancing specification GRN in the distinct cell lineages of embryos. Moreover, we found that the expression of some of the tissue-specific effector genes commences before cell lineage segregation occurs. CONCLUSIONS Based on this finding, we propose that the expression onset of tissue-specific effector genes is controlled more dynamically than suggested in the previously proposed simplistic regulation scheme. Thus, we suggest that differentiation should be conceptualized as a seamless process of accumulation of effector expression along with the advancing specification GRN. This pattern of effector gene expression may have interesting implications for the evolution of novel cell types.
Collapse
Affiliation(s)
- Shumpei Yamakawa
- Institute of Zoology and Evolutionary Research, Friedrich-Shiller University Jena, Erbertstraße 1, 07747, Jena, Germany.
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan.
| | - Atsuko Yamazaki
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Yoshiaki Morino
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Hiroshi Wada
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| |
Collapse
|
3
|
Rodríguez-Sastre N, Shapiro N, Hawkins DY, Lion AT, Peyreau M, Correa AE, Dionne K, Bradham CA. Ethanol exposure perturbs sea urchin development and disrupts developmental timing. Dev Biol 2023; 493:89-102. [PMID: 36368523 DOI: 10.1016/j.ydbio.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Ethanol is a known vertebrate teratogen that causes craniofacial defects as a component of fetal alcohol syndrome (FAS). Our results show that sea urchin embryos treated with ethanol similarly show broad skeletal patterning defects, potentially analogous to the defects associated with FAS. The sea urchin larval skeleton is a simple patterning system that involves only two cell types: the primary mesenchymal cells (PMCs) that secrete the calcium carbonate skeleton and the ectodermal cells that provide migratory, positional, and differentiation cues for the PMCs. Perturbations in RA biosynthesis and Hh signaling pathways are thought to be causal for the FAS phenotype in vertebrates. Surprisingly, our results indicate that these pathways are not functionally relevant for the teratogenic effects of ethanol in developing sea urchins. We found that developmental morphology as well as the expression of some ectodermal and PMC genes was delayed by ethanol exposure. Temporal transcriptome analysis revealed significant impacts of ethanol on signaling and metabolic gene expression, and a disruption in the timing of GRN gene expression that includes both delayed and precocious gene expression throughout the specification network. We conclude that the skeletal patterning perturbations in ethanol-treated embryos likely arise from a loss of temporal synchrony within and between the instructive and responsive tissues.
Collapse
Affiliation(s)
| | | | | | - Alexandra T Lion
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA
| | | | - Andrea E Correa
- Universidad de Puerto Rico-Recinto Aguadilla, Puerto Rico, USA
| | | | - Cynthia A Bradham
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA; Biological Design Center, Boston University, Boston, MA, USA.
| |
Collapse
|
4
|
Davidson PL, Byrne M, Wray GA. Evolutionary Changes in the Chromatin Landscape Contribute to Reorganization of a Developmental Gene Network During Rapid Life History Evolution in Sea Urchins. Mol Biol Evol 2022; 39:msac172. [PMID: 35946348 PMCID: PMC9435058 DOI: 10.1093/molbev/msac172] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Chromatin configuration is highly dynamic during embryonic development in animals, exerting an important point of control in transcriptional regulation. Yet there exists remarkably little information about the role of evolutionary changes in chromatin configuration to the evolution of gene expression and organismal traits. Genome-wide assays of chromatin configuration, coupled with whole-genome alignments, can help address this gap in knowledge in several ways. In this study we present a comparative analysis of regulatory element sequences and accessibility throughout embryogenesis in three sea urchin species with divergent life histories: a lecithotroph Heliocidaris erythrogramma, a closely related planktotroph H. tuberculata, and a distantly related planktotroph Lytechinus variegatus. We identified distinct epigenetic and mutational signatures of evolutionary modifications to the function of putative cis-regulatory elements in H. erythrogramma that have accumulated nonuniformly throughout the genome, suggesting selection, rather than drift, underlies many modifications associated with the derived life history. Specifically, regulatory elements composing the sea urchin developmental gene regulatory network are enriched for signatures of positive selection and accessibility changes which may function to alter binding affinity and access of developmental transcription factors to these sites. Furthermore, regulatory element changes often correlate with divergent expression patterns of genes involved in cell type specification, morphogenesis, and development of other derived traits, suggesting these evolutionary modifications have been consequential for phenotypic evolution in H. erythrogramma. Collectively, our results demonstrate that selective pressures imposed by changes in developmental life history rapidly reshape the cis-regulatory landscape of core developmental genes to generate novel traits and embryonic programs.
Collapse
Affiliation(s)
| | - Maria Byrne
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
5
|
Suzuki H, Yaguchi S. Direct TGF-ß signaling via alk4/5/7 pathway is involved in gut bending in sea urchin embryos. Dev Dyn 2021; 251:226-234. [PMID: 34816532 DOI: 10.1002/dvdy.442] [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: 09/15/2021] [Revised: 11/05/2021] [Accepted: 11/21/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Precise gastrulation is essential for formation of functional bodies in cnidarians and bilaterians. Previously, by using an alk4/5/7-specific inhibitor, we showed that transforming growth factor-beta (TGF-ß)-alk4/5/7 signaling pathway is important for correct gut bending in sea urchin embryos. However, it is still unclear where functional TGF-ß signals are received in embryos for correct gut bending because details of the spatiotemporal expression pattern of alk4/5/7 have not been reported. RESULTS We revealed that alk4/5/7 are expressed from the 2-cell to early pluteus stage throughout the entire body, including the invaginating gut. To investigate whether TGF-ß signals directly received in endoderm are required for correct gut bending, we made chimeras in which alk4/5/7 translation was inhibited only in endomesoderm lineage. As a result, the gut of the chimeric embryos did not bend precisely, in contrast to the control chimeras. CONCLUSION We conclude that direct TGF-ß signaling to the endoderm via alk4/5/7 pathway regulates correct gut bending. However, TGF-ß-alk4/5/7 pathway is not related to mouth opening because the mouth is formed without TGF-ß signaling to the endoderm. This research contributes to understanding the mechanisms leading to the proper positioning of the end of the archenteron for forming a through-gut, which is commonly needed for bilaterians.
Collapse
Affiliation(s)
- Haruka Suzuki
- Shimoda Marine Research Center, University of Tsukuba, Shizuoka, Japan
| | - Shunsuke Yaguchi
- Shimoda Marine Research Center, University of Tsukuba, Shizuoka, Japan.,PRESTO, JST, Kawaguchi, Saitama, Japan
| |
Collapse
|
6
|
Albarano L, Zupo V, Guida M, Libralato G, Caramiello D, Ruocco N, Costantini M. PAHs and PCBs Affect Functionally Intercorrelated Genes in the Sea Urchin Paracentrotus lividus Embryos. Int J Mol Sci 2021; 22:ijms222212498. [PMID: 34830379 PMCID: PMC8619768 DOI: 10.3390/ijms222212498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 01/05/2023] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) and polychlorinated biphenyls (PCBs) represent the most common pollutants in the marine sediments. Previous investigations demonstrated short-term sublethal effects of sediments polluted with both contaminants on the sea urchin Paracentrotus lividus after 2 months of exposure in mesocosms. In particular, morphological malformations observed in P. lividus embryos deriving from adults exposed to PAHs and PCBs were explained at molecular levels by de novo transcriptome assembly and real-time qPCR, leading to the identification of several differentially expressed genes involved in key physiological processes. Here, we extensively explored the genes involved in the response of the sea urchin P. lividus to PAHs and PCBs. Firstly, 25 new genes were identified and interactomic analysis revealed that they were functionally connected among them and to several genes previously defined as molecular targets of response to the two pollutants under analysis. The expression levels of these 25 genes were followed by Real Time qPCR, showing that almost all genes analyzed were affected by PAHs and PCBs. These findings represent an important further step in defining the impacts of slight concentrations of such contaminants on sea urchins and, more in general, on marine biota, increasing our knowledge of molecular targets involved in responses to environmental stressors.
Collapse
Affiliation(s)
- Luisa Albarano
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, Villa Comunale, 80121 Naples, Italy; (L.A.); (G.L.); (N.R.)
- Department of Biology, University of Naples Federico II, Complesso di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy;
| | - Valerio Zupo
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, Villa Dohrn, Punta San Pietro, 80077 Naples, Italy;
| | - Marco Guida
- Department of Biology, University of Naples Federico II, Complesso di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy;
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy
| | - Giovanni Libralato
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, Villa Comunale, 80121 Naples, Italy; (L.A.); (G.L.); (N.R.)
- Department of Biology, University of Naples Federico II, Complesso di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy;
| | - Davide Caramiello
- Stazione Zoologica Anton Dohrn, Department of Research Infrastructures for Marine Biological Resources, Marine Organisms Core Facility, Villa Comunale, 80121 Naples, Italy;
| | - Nadia Ruocco
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, Villa Comunale, 80121 Naples, Italy; (L.A.); (G.L.); (N.R.)
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, C. da Torre Spaccata, 87071 Amendolara, Italy
| | - Maria Costantini
- Stazione Zoologica Anton Dohrn, Department of Marine Biotechnology, Villa Comunale, 80121 Naples, Italy; (L.A.); (G.L.); (N.R.)
- Correspondence:
| |
Collapse
|
7
|
Abstract
The hedgehog (Hh) signaling pathway plays several diverse regulatory and patterning roles during organogenesis of the intestine and in the regulation of adult intestinal homeostasis. In the embryo, fetus, and adult, intestinal Hh signaling is paracrine: Hh ligands are expressed in the endodermally derived epithelium, while signal transduction is confined to the mesenchymal compartment, where at least a dozen distinct cell types are capable of responding to Hh signals. Epithelial Hh ligands not only regulate a variety of mesenchymal cell behaviors, but they also direct these mesenchymal cells to secrete additional soluble factors (e.g., Wnts, Bmps, inflammatory mediators) that feed back to regulate the epithelial cells themselves. Evolutionary conservation of the core Hh signaling pathway, as well as conservation of epithelial/mesenchymal cross talk in the intestine, has meant that work in many diverse model systems has contributed to our current understanding of the role of this pathway in intestinal organogenesis, which is reviewed here.
Collapse
Affiliation(s)
- Katherine D Walton
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA; ,
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA; ,
| |
Collapse
|
8
|
Chen CY, McKinney SA, Ellington LR, Gibson MC. Hedgehog signaling is required for endomesodermal patterning and germ cell development in the sea anemone Nematostella vectensis. eLife 2020; 9:e54573. [PMID: 32969790 PMCID: PMC7515634 DOI: 10.7554/elife.54573] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 09/05/2020] [Indexed: 12/27/2022] Open
Abstract
Two distinct mechanisms for primordial germ cell (PGC) specification are observed within Bilatera: early determination by maternal factors or late induction by zygotic cues. Here we investigate the molecular basis for PGC specification in Nematostella, a representative pre-bilaterian animal where PGCs arise as paired endomesodermal cell clusters during early development. We first present evidence that the putative PGCs delaminate from the endomesoderm upon feeding, migrate into the gonad primordia, and mature into germ cells. We then show that the PGC clusters arise at the interface between hedgehog1 and patched domains in the developing mesenteries and use gene knockdown, knockout and inhibitor experiments to demonstrate that Hh signaling is required for both PGC specification and general endomesodermal patterning. These results provide evidence that the Nematostella germline is specified by inductive signals rather than maternal factors, and support the existence of zygotically-induced PGCs in the eumetazoan common ancestor.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Sean A McKinney
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | - Matthew C Gibson
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, The University of Kansas School of MedicineKansas CityUnited States
| |
Collapse
|
9
|
Hedgehog-FGF signaling axis patterns anterior mesoderm during gastrulation. Proc Natl Acad Sci U S A 2020; 117:15712-15723. [PMID: 32561646 DOI: 10.1073/pnas.1914167117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mechanisms used by embryos to pattern tissues across their axes has fascinated developmental biologists since the founding of embryology. Here, using single-cell technology, we interrogate complex patterning defects and define a Hedgehog (Hh)-fibroblast growth factor (FGF) signaling axis required for anterior mesoderm lineage development during gastrulation. Single-cell transcriptome analysis of Hh-deficient mesoderm revealed selective deficits in anterior mesoderm populations, culminating in defects to anterior embryonic structures, including the pharyngeal arches, heart, and anterior somites. Transcriptional profiling of Hh-deficient mesoderm during gastrulation revealed disruptions to both transcriptional patterning of the mesoderm and FGF signaling for mesoderm migration. Mesoderm-specific Fgf4/Fgf8 double-mutants recapitulated anterior mesoderm defects and Hh-dependent GLI transcription factors modulated enhancers at FGF gene loci. Cellular migration defects during gastrulation induced by Hh pathway antagonism were mitigated by the addition of FGF4 protein. These findings implicate a multicomponent signaling hierarchy activated by Hh ligands from the embryonic node and executed by FGF signals in nascent mesoderm to control anterior mesoderm patterning.
Collapse
|
10
|
Gigante ED, Caspary T. Signaling in the primary cilium through the lens of the Hedgehog pathway. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 9:e377. [PMID: 32084300 DOI: 10.1002/wdev.377] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 12/14/2022]
Abstract
Cilia are microtubule-based, cell-surface projections whose machinery is evolutionarily conserved. In vertebrates, cilia are observed on almost every cell type and are either motile or immotile. Immotile sensory, or primary cilia, are responsive to extracellular ligands and signals. Cilia can be thought of as compartments, functionally distinct from the cell that provides an environment for signaling cascades. Hedgehog is a critical developmental signaling pathway which is functionally linked to primary cilia in vertebrates. The major components of the vertebrate Hedgehog signaling pathway dynamically localize to the ciliary compartment and ciliary membrane. Critically, G-protein coupled receptor (GPCR) Smoothened, the obligate transducer of the pathway, is enriched and activated in the cilium. While Smoothened is the most intensely studied ciliary receptor, many GPCRs localize within cilia. Understanding the link between Smoothened and cilia defines common features, and distinctions, of GPCR signaling within the primary cilium. This article is categorized under: Signaling Pathways > Global Signaling Mechanisms Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Eduardo D Gigante
- Graduate Program in Neuroscience, Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Hogan JD, Keenan JL, Luo L, Ibn-Salem J, Lamba A, Schatzberg D, Piacentino ML, Zuch DT, Core AB, Blumberg C, Timmermann B, Grau JH, Speranza E, Andrade-Navarro MA, Irie N, Poustka AJ, Bradham CA. The developmental transcriptome for Lytechinus variegatus exhibits temporally punctuated gene expression changes. Dev Biol 2019; 460:139-154. [PMID: 31816285 DOI: 10.1016/j.ydbio.2019.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 10/25/2022]
Abstract
Embryonic development is arguably the most complex process an organism undergoes during its lifetime, and understanding this complexity is best approached with a systems-level perspective. The sea urchin has become a highly valuable model organism for understanding developmental specification, morphogenesis, and evolution. As a non-chordate deuterostome, the sea urchin occupies an important evolutionary niche between protostomes and vertebrates. Lytechinus variegatus (Lv) is an Atlantic species that has been well studied, and which has provided important insights into signal transduction, patterning, and morphogenetic changes during embryonic and larval development. The Pacific species, Strongylocentrotus purpuratus (Sp), is another well-studied sea urchin, particularly for gene regulatory networks (GRNs) and cis-regulatory analyses. A well-annotated genome and transcriptome for Sp are available, but similar resources have not been developed for Lv. Here, we provide an analysis of the Lv transcriptome at 11 timepoints during embryonic and larval development. Temporal analysis suggests that the gene regulatory networks that underlie specification are well-conserved among sea urchin species. We show that the major transitions in variation of embryonic transcription divide the developmental time series into four distinct, temporally sequential phases. Our work shows that sea urchin development occurs via sequential intervals of relatively stable gene expression states that are punctuated by abrupt transitions.
Collapse
Affiliation(s)
- John D Hogan
- Program in Bioinformatics, Boston University, Boston, MA, USA
| | | | - Lingqi Luo
- Program in Bioinformatics, Boston University, Boston, MA, USA
| | - Jonas Ibn-Salem
- Evolution and Development Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany; Faculty of Biology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Arjun Lamba
- Biology Department, Boston University, Boston, MA, USA
| | | | - Michael L Piacentino
- Program in Molecular and Cellular Biology and Biochemistry, Boston University, Boston, MA, USA
| | - Daniel T Zuch
- Program in Molecular and Cellular Biology and Biochemistry, Boston University, Boston, MA, USA
| | - Amanda B Core
- Biology Department, Boston University, Boston, MA, USA
| | | | - Bernd Timmermann
- Sequencing Core Facility, Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - José Horacio Grau
- Dahlem Centre for Genome Research and Medical Systems Biology, Environmental and Phylogenomics Group, Berlin, Germany; Museum für Naturkunde Berlin, Leibniz-Institut für Evolutions- und Biodiversitätsforschung, Berlin, Germany
| | - Emily Speranza
- Program in Bioinformatics, Boston University, Boston, MA, USA
| | | | - Naoki Irie
- Department of Biological Sciences, University of Tokyo, Tokyo, Japan
| | - Albert J Poustka
- Evolution and Development Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany; Dahlem Centre for Genome Research and Medical Systems Biology, Environmental and Phylogenomics Group, Berlin, Germany
| | - Cynthia A Bradham
- Program in Bioinformatics, Boston University, Boston, MA, USA; Biology Department, Boston University, Boston, MA, USA; Program in Molecular and Cellular Biology and Biochemistry, Boston University, Boston, MA, USA.
| |
Collapse
|
12
|
Abstract
Gastrulation is arguably the most important evolutionary innovation in the animal kingdom. This process provides the basic embryonic architecture, an inner layer separated from an outer layer, from which all animal forms arise. An extraordinarily simple and elegant process of gastrulation is observed in the sea urchin embryo. The cells participating in sea urchin gastrulation are specified early during cleavage. One outcome of that specification is the expression of transcription factors that control each of the many subsequent morphogenetic changes. The first of these movements is an epithelial-mesenchymal transition (EMT) of skeletogenic mesenchyme cells, then EMT of pigment cell progenitors. Shortly thereafter, invagination of the archenteron occurs. At the end of archenteron extension, a second wave of EMT occurs to release immune cells into the blastocoel and primordial germ cells that will home to the coelomic pouches. The archenteron then remodels to establish the three parts of the gut, and at the anterior end, the gut fuses with the stomodaeum to form the through-gut. As part of the anterior remodeling, mesodermal coelomic pouches bud off the lateral sides of the archenteron tip. Multiple cell biological processes conduct each of these movements and in some cases the upstream transcription factors controlling this process have been identified. Remarkably, each event seamlessly occurs at the right time to orchestrate formation of the primitive body plan. This review covers progress toward understanding many of the molecular mechanisms underlying this sequence of morphogenetic events.
Collapse
|
13
|
Abstract
The adult gastrointestinal tract (GI) is a series of connected organs (esophagus, stomach, small intestine, colon) that develop via progressive regional specification of a continuous tubular embryonic organ anlage. This chapter focuses on organogenesis of the small intestine. The intestine arises by folding of a flat sheet of endodermal cells into a tube of highly proliferative pseudostratified cells. Dramatic elongation of this tube is driven by rapid epithelial proliferation. Then, epithelial-mesenchymal crosstalk and physical forces drive a stepwise cascade that results in convolution of the tubular surface into finger-like projections called villi. Concomitant with villus formation, a sharp epithelial transcriptional boundary is defined between stomach and intestine. Finally, flask-like depressions called crypts are established to house the intestinal stem cells needed throughout life for epithelial renewal. New insights into these events are being provided by in vitro organoid systems, which hold promise for future regenerative engineering of the small intestine.
Collapse
Affiliation(s)
- Sha Wang
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| | - Katherine D Walton
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States.
| | - Deborah L Gumucio
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| |
Collapse
|
14
|
Adachi S, Niimi I, Sakai Y, Sato F, Minokawa T, Urata M, Sehara-Fujisawa A, Kobayashi I, Yamaguchi M. Anteroposterior molecular registries in ectoderm of the echinus rudiment. Dev Dyn 2018; 247:1297-1307. [DOI: 10.1002/dvdy.24686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/03/2018] [Accepted: 10/21/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Shinya Adachi
- Graduate School of Natural Science and Technology; Kanazawa University; Kakuma Kanazawa Japan
| | - Iyo Niimi
- Graduate School of Natural Science and Technology; Kanazawa University; Kakuma Kanazawa Japan
| | - Yui Sakai
- Graduate School of Natural Science and Technology; Kanazawa University; Kakuma Kanazawa Japan
| | - Fuminori Sato
- Department of Growth Regulation; Institute for Frontier Medical Sciences, Kyoto University; Sakyo-ku Kyoto Japan
| | - Takuya Minokawa
- Research Center for Marine Biology, Graduate School of Life Sciences; Tohoku University; Asamushi Aomori Japan
| | - Makoto Urata
- Noto Marine Laboratory, Institute of Natural and Environmental Technology; Kanazawa University; Noto Hosu Japan
| | - Atsuko Sehara-Fujisawa
- Department of Growth Regulation; Institute for Frontier Medical Sciences, Kyoto University; Sakyo-ku Kyoto Japan
| | - Isao Kobayashi
- Graduate School of Natural Science and Technology; Kanazawa University; Kakuma Kanazawa Japan
| | - Masaaki Yamaguchi
- Graduate School of Natural Science and Technology; Kanazawa University; Kakuma Kanazawa Japan
| |
Collapse
|
15
|
Cui M, Lin CY, Su YH. Recent advances in functional perturbation and genome editing techniques in studying sea urchin development. Brief Funct Genomics 2018; 16:309-318. [PMID: 28605407 DOI: 10.1093/bfgp/elx011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Studies on the gene regulatory networks (GRNs) of sea urchin embryos have provided a basic understanding of the molecular mechanisms controlling animal development. The causal links in GRNs have been verified experimentally through perturbation of gene functions. Microinjection of antisense morpholino oligonucleotides (MOs) into the egg is the most widely used approach for gene knockdown in sea urchin embryos. The modification of MOs into a membrane-permeable form (vivo-MOs) has allowed gene knockdown at later developmental stages. Recent advances in genome editing tools, such as zinc-finger nucleases, transcription activator-like effector-based nucleases and the clustered regularly interspaced short palindromic repeat/clustered regularly interspaced short palindromic repeat-associated protein 9 (CRISPR/Cas9) system, have provided methods for gene knockout in sea urchins. Here, we review the use of vivo-MOs and genome editing tools in sea urchin studies since the publication of its genome in 2006. Various applications of the CRISPR/Cas9 system and their potential in studying sea urchin development are also discussed. These new tools will provide more sophisticated experimental methods for studying sea urchin development.
Collapse
|
16
|
Zhang X, Chen Y, Ye Y, Wang J, Wang H, Yuan G, Lin Z, Wu Y, Zhang Y, Lin X. Wnt signaling promotes hindgut fate commitment through regulating multi-lineage genes during hESC differentiation. Cell Signal 2016; 29:12-22. [PMID: 27693749 DOI: 10.1016/j.cellsig.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Wnt signaling plays essential roles in both embryonic pattern formation and postembryonic tissue homoestasis. High levels of Wnt activity repress foregut identity and facilitate hindgut fate through forming a gradient of Wnt signaling activity along the anterior-posterior axis. Here, we examined the mechanisms of Wnt signaling in hindgut development by differentiating human embryonic stem cells (hESCs) into the hindgut progenitors. We observed severe morphological changes when Wnt signaling was blocked by using Wnt antagonist Dkk1. We performed deep-transcriptome sequencing (RNA-seq) and identified 240 Wnt-activated genes and 2023 Wnt-repressed genes, respectively. Clusters of Wnt targets showed enrichment in specific biological functions, such as "gastrointestinal or skeletal development" in the Wnt-activated targets and "neural or immune system development" in the Wnt-repressed targets. Moreover, we adopted a high-throughput chromatin immunoprecipitation and deep sequencing (ChIP-seq) approach to identify the genomic regions through which Wnt-activated transcription factor TCF7L2 regulated transcription. We identified 83 Wnt direct target candidates, including the hindgut marker CDX2 and the genes relevant to morphogenesis (MSX1, MSX2, LEF1, T, PDGFRB etc.) through combinatorial analysis of the RNA-seq and ChIP-seq data. Together, our study identified a series of direct and indirect Wnt targets in hindgut differentiation, and uncovered the diverse mechanisms of Wnt signaling in regulating multi-lineage differentiation.
Collapse
Affiliation(s)
- Xiujuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying Ye
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jianfeng Wang
- Core Genomic Facility, CAS Key Laboratory of Genome Sciences & Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Hong Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guohong Yuan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yihui Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinhua Lin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Division of Developmental Biology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Piacentino ML, Chung O, Ramachandran J, Zuch DT, Yu J, Conaway EA, Reyna AE, Bradham CA. Zygotic LvBMP5-8 is required for skeletal patterning and for left–right but not dorsal–ventral specification in the sea urchin embryo. Dev Biol 2016; 412:44-56. [DOI: 10.1016/j.ydbio.2016.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/31/2016] [Accepted: 02/18/2016] [Indexed: 01/25/2023]
|
18
|
Martik ML, Lyons DC, McClay DR. Developmental gene regulatory networks in sea urchins and what we can learn from them. F1000Res 2016; 5. [PMID: 26962438 PMCID: PMC4765714 DOI: 10.12688/f1000research.7381.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2016] [Indexed: 12/21/2022] Open
Abstract
Sea urchin embryos begin zygotic transcription shortly after the egg is fertilized. Throughout the cleavage stages a series of transcription factors are activated and, along with signaling through a number of pathways, at least 15 different cell types are specified by the beginning of gastrulation. Experimentally, perturbation of contributing transcription factors, signals and receptors and their molecular consequences enabled the assembly of an extensive gene regulatory network model. That effort, pioneered and led by Eric Davidson and his laboratory, with many additional insights provided by other laboratories, provided the sea urchin community with a valuable resource. Here we describe the approaches used to enable the assembly of an advanced gene regulatory network model describing molecular diversification during early development. We then provide examples to show how a relatively advanced authenticated network can be used as a tool for discovery of how diverse developmental mechanisms are controlled and work.
Collapse
Affiliation(s)
- Megan L Martik
- Biology Department, Duke University, Durham, North Carolina, 27708, USA
| | - Deirdre C Lyons
- Biology Department, Duke University, Durham, North Carolina, 27708, USA
| | - David R McClay
- Biology Department, Duke University, Durham, North Carolina, 27708, USA
| |
Collapse
|
19
|
Warner JF, Miranda EL, McClay DR. Contribution of hedgehog signaling to the establishment of left-right asymmetry in the sea urchin. Dev Biol 2016; 411:314-324. [PMID: 26872875 DOI: 10.1016/j.ydbio.2016.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 02/06/2016] [Accepted: 02/08/2016] [Indexed: 01/22/2023]
Abstract
Most bilaterians exhibit a left-right asymmetric distribution of their internal organs. The sea urchin larva is notable in this regard since most adult structures are generated from left sided embryonic structures. The gene regulatory network governing this larval asymmetry is still a work in progress but involves several conserved signaling pathways including Nodal, and BMP. Here we provide a comprehensive analysis of Hedgehog signaling and it's contribution to left-right asymmetry. We report that Hh signaling plays a conserved role to regulate late asymmetric expression of Nodal and that this regulation occurs after Nodal breaks left-right symmetry in the mesoderm. Thus, while Hh functions to maintain late Nodal expression, the molecular asymmetry of the future coelomic pouches is locked in. Furthermore we report that cilia play a role only insofar as to transduce Hh signaling and do not have an independent effect on the asymmetry of the mesoderm. From this, we are able to construct a more complete regulatory network governing the establishment of left-right asymmetry in the sea urchin.
Collapse
Affiliation(s)
- Jacob F Warner
- Duke University Department of Biology, Durham, NC, United States
| | - Esther L Miranda
- Duke University Department of Biology, Durham, NC, United States
| | - David R McClay
- Duke University Department of Biology, Durham, NC, United States.
| |
Collapse
|
20
|
Schatzberg D, Lawton M, Hadyniak SE, Ross EJ, Carney T, Beane WS, Levin M, Bradham CA. H(+)/K(+) ATPase activity is required for biomineralization in sea urchin embryos. Dev Biol 2015; 406:259-70. [PMID: 26282894 DOI: 10.1016/j.ydbio.2015.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/26/2015] [Accepted: 08/13/2015] [Indexed: 12/31/2022]
Abstract
The bioelectrical signatures associated with regeneration, wound healing, development, and cancer are changes in the polarization state of the cell that persist over long durations, and are mediated by ion channel activity. To identify physiologically relevant bioelectrical changes that occur during normal development of the sea urchin Lytechinus variegatus, we tested a range of ion channel inhibitors, and thereby identified SCH28080, a chemical inhibitor of the H(+)/K(+) ATPase (HKA), as an inhibitor of skeletogenesis. In sea urchin embryos, the primary mesodermal lineage, the PMCs, produce biomineral in response to signals from the ectoderm. However, in SCH28080-treated embryos, aside from randomization of the left-right axis, the ectoderm is normally specified and differentiated, indicating that the block to skeletogenesis observed in SCH28080-treated embryos is PMC-specific. HKA inhibition did not interfere with PMC specification, and was sufficient to block continuing biomineralization when embryos were treated with SCH28080 after the initiation of skeletogenesis, indicating that HKA activity is continuously required during biomineralization. Ion concentrations and voltage potential were abnormal in the PMCs in SCH28080-treated embryos, suggesting that these bioelectrical abnormalities prevent biomineralization. Our results indicate that this effect is due to the inhibition of amorphous calcium carbonate precipitation within PMC vesicles.
Collapse
Affiliation(s)
| | - Matthew Lawton
- Department of Biology, Boston University, Boston, MA 02215, USA
| | | | - Erik J Ross
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Tamara Carney
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Wendy S Beane
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA 02155, USA
| | | |
Collapse
|
21
|
Andrikou C, Pai CY, Su YH, Arnone MI. Logics and properties of a genetic regulatory program that drives embryonic muscle development in an echinoderm. eLife 2015. [PMID: 26218224 PMCID: PMC4549668 DOI: 10.7554/elife.07343] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Evolutionary origin of muscle is a central question when discussing mesoderm evolution. Developmental mechanisms underlying somatic muscle development have mostly been studied in vertebrates and fly where multiple signals and hierarchic genetic regulatory cascades selectively specify myoblasts from a pool of naive mesodermal progenitors. However, due to the increased organismic complexity and distant phylogenetic position of the two systems, a general mechanistic understanding of myogenesis is still lacking. In this study, we propose a gene regulatory network (GRN) model that promotes myogenesis in the sea urchin embryo, an early branching deuterostome. A fibroblast growth factor signaling and four Forkhead transcription factors consist the central part of our model and appear to orchestrate the myogenic process. The topological properties of the network reveal dense gene interwiring and a multilevel transcriptional regulation of conserved and novel myogenic genes. Finally, the comparison of the myogenic network architecture among different animal groups highlights the evolutionary plasticity of developmental GRNs. DOI:http://dx.doi.org/10.7554/eLife.07343.001 Muscles, bones, and blood vessels all develop from a tissue called the mesoderm, which forms early on in the development of an embryo. Networks of genes control which parts of the mesoderm transform into different cell types. The gene networks that control the development of muscle cells from the mesoderm have so far been investigated in flies and several species of animals with backbones. However, these species are complex, which makes it difficult to work out the general principles that control muscle cell development. Sea urchins are often studied in developmental biology as they have many of the same genes as more complex animals, but are much simpler and easier to study in the laboratory. Andrikou et al. therefore investigated the ‘gene regulatory network’ that controls muscle development in sea urchins. This revealed that proteins called Forkhead transcription factors and a process called FGF signaling are crucial for controlling muscle development in sea urchins. These are also important factors for developing muscles in other animals. Andrikou et al. then produced models that show the interactions between the genes that control muscle formation at three different stages of embryonic development. These models reveal several important features of the muscle development gene regulatory network. For example, the network is robust: if one gene fails, the network is connected in a way that allows it to still make muscle. This also allows the network to adapt and evolve without losing the ability to perform any of its existing roles. Comparing the gene regulatory network that controls muscle development in sea urchins with the networks found in other animals showed that many of the same genes are used across different species, but are connected into different network structures. Investigating the similarities and differences of the regulatory networks in different species could help us to understand how muscles have evolved and could ultimately lead to a better understanding of the causes of developmental diseases. DOI:http://dx.doi.org/10.7554/eLife.07343.002
Collapse
Affiliation(s)
- Carmen Andrikou
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Chih-Yu Pai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Hsien Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Maria Ina Arnone
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Napoli, Italy
| |
Collapse
|
22
|
|
23
|
Piacentino ML, Ramachandran J, Bradham CA. Late Alk4/5/7 signaling is required for anterior skeletal patterning in sea urchin embryos. Development 2015; 142:943-52. [PMID: 25633352 DOI: 10.1242/dev.114322] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Skeletal patterning in the sea urchin embryo requires a conversation between the skeletogenic primary mesenchyme cells (PMCs) and the overlying pattern-dictating ectoderm; however, our understanding of the molecular basis for this process remains incomplete. Here, we show that TGF-β-receptor signaling is required during gastrulation to pattern the anterior skeleton. To block TGF-β signaling, we used SB431542 (SB43), a specific inhibitor of the TGF-β type I receptor Alk4/5/7. Treatment with SB43 during gastrulation blocks anterior PMC positioning and the formation of the anterior skeleton, but does not perturb general ectoderm specification or development. This is the first example of a signaling event required for patterning of a specific part of the skeleton. Alk4/5/7 inhibition does not prevent the formation of a mouth, although SB43-treated plutei display reduced feeding ability, presumably due to the loss of the structural support for the mouth conferred by the anterior skeleton. Both Univin and Nodal are potential ligands for Alk4/5/7; however, Nodal is unilaterally expressed on only the right side, whereas Univin is bilaterally expressed in the ectoderm adjacent to the anterior skeleton during the relevant time period. Our results demonstrate that Univin is both necessary and sufficient for secondary skeletal development in a control background, consistent with the hypothesis that Univin is a relevant Alk4/5/7 ligand for anterior skeletal patterning. Taken together, our data demonstrate that Alk4/5/7 signaling during gastrulation is required to direct PMCs to the oral hood, and suggest that Univin is a relevant ligand for this signaling event.
Collapse
Affiliation(s)
- Michael L Piacentino
- Department of Biology, Boston University, Boston, MA 02215, USA Program in Molecular Biology, Cell Biology and Biochemistry, Boston University, Boston, MA 02215, USA
| | | | - Cynthia A Bradham
- Department of Biology, Boston University, Boston, MA 02215, USA Program in Molecular Biology, Cell Biology and Biochemistry, Boston University, Boston, MA 02215, USA Program in Bioinformatics, Boston University, Boston, MA 02215, USA
| |
Collapse
|
24
|
Abstract
The Hedgehog pathway has been shown to be an important developmental signaling pathway in many organisms (Ingham and McMahon. Genes Dev 15:3059-3087, 2001). Recently that work has been extended to developing echinoderm embryos (Walton et al. Dev Biol 331(1):26-37, 2009). Here we describe several methods to perturb the Hedgehog signaling pathway in the sea urchin. These include microinjection of Morpholinos and mRNA constructs as well as treatments with small molecule inhibitors. Finally we provide simple methods for assaying Hedgehog phenotypes in the sea urchin embryo.
Collapse
|
25
|
Byrum CA, Smith J, Easterling MR, Bridges MC. Restricted expression of karyopherin alpha mRNA in the sea urchin suggests a role in neurogenesis. Gene Expr Patterns 2014; 16:51-60. [PMID: 25218279 DOI: 10.1016/j.gep.2014.06.005] [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: 10/07/2013] [Revised: 06/11/2014] [Accepted: 06/25/2014] [Indexed: 10/24/2022]
Abstract
Karyopherin alpha (KAP-α) proteins are critical for the transport of many molecules into the nucleus. In this study, we identified three members of the KAP-α family in the sea urchin Lytechinus variegatus and described the developmental expression of these proteins. Although many importins are assumed to have ubiquitous expression, we found that all three genes were differentially expressed. Both LvKPNA1/5/6 and LvKPNA3/4 accumulated at high levels during cleavage, exhibiting cyclic expression as cells divided. By the blastula and gastrula stages expression decreased, remaining highest in the vegetal plate and archenteron, and by the prism/pluteus stages expression was restricted to the oral surface and gut. Expression of a third KAP-α gene, LvKPNA2/7, was examined in embryos from the mesenchyme blastula to pluteus stages. LvKPNA2/7 mRNA is present in vegetal cells of the mesenchyme blastula and, during gastrulation, it is localized to the archenteron and appears in additional groups of ectodermal cells. Prism/pluteus stage embryos expressed LvKPNA2/7 in the gut and scattered distribution of transcripts in the ciliary band resembled expression patterns of neural cells. We hypothesize that LvKPNA2/7 maintains pluripotency in the neural precursors prior to activation of neural differentiation and believe that this study is an important first step in an effort to better understand the roles of importins during embryogenesis.
Collapse
Affiliation(s)
- Christine A Byrum
- Department of Biology, College of Charleston, Rita Liddy Hollings Science Center, 58 Coming Street, Room 214, Charleston, SC, USA.
| | - Jason Smith
- Department of Biology, College of Charleston, Rita Liddy Hollings Science Center, 58 Coming Street, Room 214, Charleston, SC, USA
| | - Marietta R Easterling
- Department of Biology, College of Charleston, Rita Liddy Hollings Science Center, 58 Coming Street, Room 214, Charleston, SC, USA
| | - M Catherine Bridges
- Department of Biology, College of Charleston, Rita Liddy Hollings Science Center, 58 Coming Street, Room 214, Charleston, SC, USA
| |
Collapse
|
26
|
Affiliation(s)
- Jacob F. Warner
- Department of Biology; Duke University; Durham North Carolina
| | - David R. McClay
- Department of Biology; Duke University; Durham North Carolina
| |
Collapse
|
27
|
Andrikou C, Iovene E, Rizzo F, Oliveri P, Arnone MI. Myogenesis in the sea urchin embryo: the molecular fingerprint of the myoblast precursors. EvoDevo 2013; 4:33. [PMID: 24295205 PMCID: PMC4175510 DOI: 10.1186/2041-9139-4-33] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/02/2013] [Indexed: 01/01/2023] Open
Abstract
Background In sea urchin larvae the circumesophageal fibers form a prominent muscle system of mesodermal origin. Although the morphology and later development of this muscle system has been well-described, little is known about the molecular signature of these cells or their precise origin in the early embryo. As an invertebrate deuterostome that is more closely related to the vertebrates than other commonly used model systems in myogenesis, the sea urchin fills an important phylogenetic gap and provides a unique perspective on the evolution of muscle cell development. Results Here, we present a comprehensive description of the development of the sea urchin larval circumesophageal muscle lineage beginning with its mesodermal origin using high-resolution localization of the expression of several myogenic transcriptional regulators and differentiation genes. A few myoblasts are bilaterally distributed at the oral vegetal side of the tip of the archenteron and first appear at the late gastrula stage. The expression of the differentiation genes Myosin Heavy Chain, Tropomyosin I and II, as well as the regulatory genes MyoD2, FoxF, FoxC, FoxL1, Myocardin, Twist, and Tbx6 uniquely identify these cells. Interestingly, evolutionarily conserved myogenic factors such as Mef2, MyoR and Six1/2 are not expressed in sea urchin myoblasts but are found in other mesodermal domains of the tip of the archenteron. The regulatory states of these domains were characterized in detail. Moreover, using a combinatorial analysis of gene expression we followed the development of the FoxF/FoxC positive cells from the onset of expression to the end of gastrulation. Our data allowed us to build a complete map of the Non-Skeletogenic Mesoderm at the very early gastrula stage, in which specific molecular signatures identify the precursors of different cell types. Among them, a small group of cells within the FoxY domain, which also express FoxC and SoxE, have been identified as plausible myoblast precursors. Together, these data support a very early gastrula stage segregation of the myogenic lineage. Conclusions From this analysis, we are able to precisely define the regulatory and differentiation signatures of the circumesophageal muscle in the sea urchin embryo. Our findings have important implications in understanding the evolution of development of the muscle cell lineage at the molecular level. The data presented here suggest a high level of conservation of the myogenic specification mechanisms across wide phylogenetic distances, but also reveal clear cases of gene cooption.
Collapse
Affiliation(s)
| | | | | | | | - Maria Ina Arnone
- Cellular and Developmental Biology, Stazione Zoologica Anton Dohrn, Napoli 80121, Italy.
| |
Collapse
|
28
|
Warner JF, McCarthy AM, Morris RL, McClay DR. Hedgehog signaling requires motile cilia in the sea urchin. Mol Biol Evol 2013; 31:18-22. [PMID: 24124205 DOI: 10.1093/molbev/mst176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A relatively small number of signaling pathways govern the early patterning processes of metazoan development. The architectural changes over time to these signaling pathways offer unique insights into their evolution. In the case of Hedgehog (Hh) signaling, two very divergent mechanisms of pathway transduction have evolved. In vertebrates, signaling relies on trafficking of Hh pathway components to nonmotile specialized primary cilia. In contrast, protostomes do not use cilia of any kind for Hh signal transduction. How these divergent lineages adapted such dramatically different ways of activating the signaling pathway is an unanswered question. Here, we present evidence that in the sea urchin, a basal deuterostome, motile cilia are required for embryonic Hh signal transduction, and the Hh receptor Smoothened (Smo) localizes to cilia during active Hh signaling. This is the first evidence that Hh signaling requires motile cilia and the first case of an organism requiring cilia outside of the vertebrate lineage.
Collapse
Affiliation(s)
- Jacob F Warner
- Duke University, University Program in Genetics and Genomics
| | | | | | | |
Collapse
|
29
|
Sławińska A, Brzezińska J, Siwek M, Elminowska-Wenda G. Expression of myogenic genes in chickens stimulated in ovo with light and temperature. Reprod Biol 2013; 13:161-5. [PMID: 23719122 DOI: 10.1016/j.repbio.2013.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/12/2012] [Indexed: 11/29/2022]
Abstract
Since chicken myogenesis is tightly controlled by myogenic regulatory factors (MRFs), the external stimuli (e.g. light or temperature) affecting the proliferation and differentiation of the muscle cells have a primary effect on the gene expression of MRFs. The aim of this study was to analyze the expression of some of MRF genes (MyoD1, myogenin and Myf5) in response to the stimulation of chicken embryos with green light (AL group) or increased temperature (38.5°C; AT group) on day 18 of embryo development (18ED) as well as on days 4 (4PHD) and 8 (8PHD) post hatch. To achieve this goal a quantitative reverse transcription polymerase chain reaction was used. The most prominent differences in gene expression were observed before hatching. Relative expression of MyoD1 on 18ED was higher (p<0.05) in AL and control (AC) groups in comparison to the AT group. Myogenin expression on 18ED was lower (p<0.05) in control chickens than in both treated groups. Light stimulation in ovo decreased (p<0.05) the Myf5 expression on 18ED in comparison to the control group. Green-light illumination applied during in ovo development had more pronounced effects on mRNA level of MRFs genes measured during both the pre- and post hatch development. The elevated temperature applied during embryonic development affected only the 18ED time point. This suggests that the effect of green-light illumination on chicken myogenesis was more prolonged than that of elevated temperature.
Collapse
Affiliation(s)
- Anna Sławińska
- University of Technology and Life Sciences in Bydgoszcz, Faculty of Animal Breeding and Biology, Department of Animal Biotechnology, 85-225 Bydgoszcz, Poland.
| | | | | | | |
Collapse
|
30
|
Materna SC, Swartz SZ, Smith J. Notch and Nodal control forkhead factor expression in the specification of multipotent progenitors in sea urchin. Development 2013; 140:1796-806. [PMID: 23533178 PMCID: PMC3621494 DOI: 10.1242/dev.091157] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2013] [Indexed: 01/23/2023]
Abstract
Indirect development, in which embryogenesis gives rise to a larval form, requires that some cells retain developmental potency until they contribute to the different tissues in the adult, including the germ line, in a later, post-embryonic phase. In sea urchins, the coelomic pouches are the major contributor to the adult, but how coelomic pouch cells (CPCs) are specified during embryogenesis is unknown. Here we identify the key signaling inputs into the CPC specification network and show that the forkhead factor foxY is the first transcription factor specifically expressed in CPC progenitors. Through dissection of its cis-regulatory apparatus we determine that the foxY expression pattern is the result of two signaling inputs: first, Delta/Notch signaling activates foxY in CPC progenitors; second, Nodal signaling restricts its expression to the left side, where the adult rudiment will form, through direct repression by the Nodal target pitx2. A third signal, Hedgehog, is required for coelomic pouch morphogenesis and institution of laterality, but does not directly affect foxY transcription. Knockdown of foxY results in a failure to form coelomic pouches and disrupts the expression of virtually all transcription factors known to be expressed in this cell type. Our experiments place foxY at the top of the regulatory hierarchy underlying the specification of a cell type that maintains developmental potency.
Collapse
Affiliation(s)
- Stefan C. Materna
- California Institute of Technology, Division of Biology, m/c 156-29, Pasadena, CA 91125, USA
| | - S. Zachary Swartz
- Brown University, Department of Molecular Biology, Cell Biology and Biochemistry, 185 Meeting Street, Providence, RI 02912, USA
| | - Joel Smith
- California Institute of Technology, Division of Biology, m/c 156-29, Pasadena, CA 91125, USA
- Brown University, Department of Molecular Biology, Cell Biology and Biochemistry, 185 Meeting Street, Providence, RI 02912, USA
- Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA 02543, USA
| |
Collapse
|
31
|
Materna SC, Ransick A, Li E, Davidson EH. Diversification of oral and aboral mesodermal regulatory states in pregastrular sea urchin embryos. Dev Biol 2012; 375:92-104. [PMID: 23261933 DOI: 10.1016/j.ydbio.2012.11.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/29/2012] [Accepted: 11/29/2012] [Indexed: 01/08/2023]
Abstract
Specification of the non-skeletogenic mesoderm (NSM) in sea urchin embryos depends on Delta signaling. Signal reception leads to expression of regulatory genes that later contribute to the aboral NSM regulatory state. In oral NSM, this is replaced by a distinct oral regulatory state in consequence of Nodal signaling. Through regulome wide analysis we identify the homeobox gene not as an immediate Nodal target. not expression in NSM causes extinction of the aboral regulatory state in the oral NSM, and expression of a new suite of regulatory genes. All NSM specific regulatory genes are henceforth expressed exclusively, in oral or aboral domains, presaging the mesodermal cell types that will emerge. We have analyzed the regulatory linkages within the aboral NSM gene regulatory network. A linchpin of this network is gataE which as we show is a direct Gcm target and part of a feedback loop locking down the aboral regulatory state.
Collapse
Affiliation(s)
- Stefan C Materna
- California Institute of Technology, Division of Biology, m/c 156-29, Pasadena, CA 91125, USA
| | | | | | | |
Collapse
|
32
|
Comparative genomics of the Hedgehog loci in chordates and the origins of Shh regulatory novelties. Sci Rep 2012; 2:433. [PMID: 22666536 PMCID: PMC3364491 DOI: 10.1038/srep00433] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 05/15/2012] [Indexed: 12/04/2022] Open
Abstract
The origin and evolution of the complex regulatory landscapes of some vertebrate developmental genes, often spanning hundreds of Kbp and including neighboring genes, remain poorly understood. The Sonic Hedgehog (Shh) genomic regulatory block (GRB) is one of the best functionally characterized examples, with several discrete enhancers reported within its introns, vast upstream gene-free region and neighboring genes (Lmbr1 and Rnf32). To investigate the origin and evolution of this GRB, we sequenced and characterized the Hedgehog (Hh) loci from three invertebrate chordate amphioxus species, which share several early expression domains with Shh. Using phylogenetic footprinting within and between chordate lineages, and reporter assays in zebrafish probing >30 Kbp of amphioxus Hh, we report large sequence and functional divergence between both groups. In addition, we show that the linkage of Shh to Lmbr1 and Rnf32, necessary for the unique gnatostomate-specific Shh limb expression, is a vertebrate novelty occurred between the two whole-genome duplications.
Collapse
|
33
|
Abstract
Embryos of the echinoderms, especially those of sea urchins and sea stars, have been studied as model organisms for over 100 years. The simplicity of their early development, and the ease of experimentally perturbing this development, provides an excellent platform for mechanistic studies of cell specification and morphogenesis. As a result, echinoderms have contributed significantly to our understanding of many developmental mechanisms, including those that govern the structure and design of gene regulatory networks, those that direct cell lineage specification, and those that regulate the dynamic morphogenetic events that shape the early embryo.
Collapse
Affiliation(s)
- David R McClay
- Department of Biology, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
34
|
Ingham PW, Nakano Y, Seger C. Mechanisms and functions of Hedgehog signalling across the metazoa. Nat Rev Genet 2011; 12:393-406. [PMID: 21502959 DOI: 10.1038/nrg2984] [Citation(s) in RCA: 446] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hedgehog proteins constitute one of a small number of families of secreted signals that have a central role in the development of metazoans. Genetic analyses in flies, fish and mice have uncovered the major components of the pathway that transduces Hedgehog signals, and recent genome sequence projects have provided clues about its evolutionary origins. In this Review we provide an updated overview of the mechanisms and functions of this signalling pathway, highlighting the conserved and divergent features of the pathway, as well as some of the common themes in its deployment that have emerged from recent studies.
Collapse
Affiliation(s)
- Philip W Ingham
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore.
| | | | | |
Collapse
|
35
|
Islam AFMT, Moly PK, Miyamoto Y, Kusakabe TG. Distinctive expression patterns of Hedgehog pathway genes in the Ciona intestinalis larva: implications for a role of Hedgehog signaling in postembryonic development and chordate evolution. Zoolog Sci 2010; 27:84-90. [PMID: 20141412 DOI: 10.2108/zsj.27.84] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Members of the Hedgehog (Hh) family are soluble ligands that orchestrate a wide spectrum of developmental processes ranging from left-right axis determination of the embryo to tissue patterning and organogenesis. Tunicates, including ascidians, are the closest relatives of vertebrates, and elucidation of Hh signaling in ascidians should provide an important clue towards better understanding the role of this pathway in development. In previous studies, expression patterns of genes encoding Hh and its downstream factor Gli have been examined up to the tailbud stage in the ascidian embryo, but their expression in the larva has not been reported. Here we show the spatial expression patterns of hedgehog (Ci-hh1, Ci-hh2), patched (Ci-ptc), smoothened (Ci-smo), and Gli (Ci-Gli) orthologs in larvae of the ascidian Ciona intestinalis. The expression patterns of Ci-hh2 and Ci-Gli dramatically change during the period between the late tailbud embryo and the swimming larva. At the larval stage, expression of Ci-Gli was found in a central part of the endoderm and in the visceral ganglion, while Ci-hh2 was expressed in two discrete endodermal regions, anteriorly and posteriorly adjacent to the cells expressing Gli. The expression patterns of these genes suggest that the Hh ligand controls postembryonic development of the endoderm and the central nervous system. Expression of a gene encoding Hh in the anterior and/or pharyngeal endoderm is probably an ancient chordate character; diversification of regulation and targets of the Hh signaling in this region may have played a major role in the evolution of chordate body structures.
Collapse
|