1
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
2
|
Abou Azar F, Lim GE. Metabolic Contributions of Wnt Signaling: More Than Controlling Flight. Front Cell Dev Biol 2021; 9:709823. [PMID: 34568323 PMCID: PMC8458764 DOI: 10.3389/fcell.2021.709823] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
The canonical Wnt signaling pathway is ubiquitous throughout the body and influences a diverse array of physiological processes. Following the initial discovery of the Wnt signaling pathway during wing development in Drosophila melanogaster, it is now widely appreciated that active Wnt signaling in mammals is necessary for the development and growth of various tissues involved in whole-body metabolism, such as brain, liver, pancreas, muscle, and adipose. Moreover, elegant gain- and loss-of-function studies have dissected the tissue-specific roles of various downstream effector molecules in the regulation of energy homeostasis. This review attempts to highlight and summarize the contributions of the Wnt signaling pathway and its downstream effectors on whole-body metabolism and their influence on the development of metabolic diseases, such as diabetes and obesity. A better understanding of the Wnt signaling pathway in these tissues may aid in guiding the development of future therapeutics to treat metabolic diseases.
Collapse
Affiliation(s)
- Frederic Abou Azar
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
3
|
Kreis J, Wielath FM, Vick P. Rab7 is required for mesoderm patterning and gastrulation in Xenopus. Biol Open 2021; 10:269049. [PMID: 34096568 PMCID: PMC8325926 DOI: 10.1242/bio.056887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/26/2021] [Indexed: 11/20/2022] Open
Abstract
Early embryogenesis requires tightly controlled temporal and spatial coordination of cellular behavior and signaling. Modulations are achieved at multiple levels, from cellular transcription to tissue-scale behavior. Intracellularly, the endolysosomal system emerges as an important regulator at different levels, but in vivo studies are rare. In the frog Xenopus, little is known about the developmental roles of endosomal regulators, or their potential involvement in signaling, especially for late endosomes. Here, we analyzed a hypothesized role of Rab7 in this context, a small GTPase known for its role as a late endosomal regulator. First, rab7 showed strong maternal expression. Following localized zygotic transcript enrichment in the mesodermal ring and neural plate, it was found in tailbud-stage neural ectoderm, notochord, pronephros, eyes and neural crest tissues. Inhibition resulted in strong axis defects caused by a requirement of rab7 for mesodermal patterning and correct gastrulation movements. To test a potential involvement in growth factor signaling, we analyzed early Wnt-dependent processes in the mesoderm. Our results suggest a selective requirement for ligand-induced Wnt activation, implicating a context-dependent role of Rab7. Summary: The late endosomal regulator Rab7 is required for gastrulation movements and axis elongation in Xenopus by regulating early mesoderm patterning.
Collapse
Affiliation(s)
- Jennifer Kreis
- Department of Zoology, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Fee M Wielath
- Department of Zoology, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Philipp Vick
- Department of Zoology, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
4
|
Félix LM, Luzio A, Themudo M, Antunes L, Matos M, Coimbra AM, Valentim AM. MS-222 short exposure induces developmental and behavioural alterations in zebrafish embryos. Reprod Toxicol 2018; 81:122-131. [DOI: 10.1016/j.reprotox.2018.07.086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 01/19/2023]
|
5
|
Li YL, Shao M, Shi DL. Rac1 signalling coordinates epiboly movement by differential regulation of actin cytoskeleton in zebrafish. Biochem Biophys Res Commun 2017; 490:1059-1065. [PMID: 28668387 DOI: 10.1016/j.bbrc.2017.06.165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 10/24/2022]
Abstract
Dynamic cytoskeleton organization is essential for polarized cell behaviours in a wide variety of morphogenetic events. In zebrafish, epiboly involves coordinated cell shape changes and expansion of cell layers to close the blastopore, but many important regulatory aspects are still unclear. Especially, the spatio-temporal regulation and function of actin structures remain to be determined for a better understanding of the mechanisms that coordinate epiboly movement. Here we show that Rac1 signalling, likely functions downstream of phosphatiditylinositol-3 kinase, is required for F-actin organization during epiboly progression in zebtafish. Using a dominant negative mutant of Rac1 and specific inhibitors to block the activation of this pathway, we find that marginal contractile actin ring is sensitive to inhibition of Rac1 signalling. In particular, we identify a novel function for this actin structure in retaining the external yolk syncytial nuclei within the margin of enveloping layer for coordinated movement toward the vegetal pole. Furthermore, we find that F-actin bundles, progressively formed in the vegetal cortex of the yolk cell, act in concert with marginal actin ring and play an active role in pulling external yolk syncytial nuclei toward the vegetal pole direction. This study uncovers novel roles of different actin structures in orchestrating epiboly movement. It helps to provide insight into the mechanisms regulating cellular polarization during early development.
Collapse
Affiliation(s)
- Yu-Long Li
- School of Life Sciences, Shandong University, 27, Shanda Nan Road, Jinan 250100, China
| | - Ming Shao
- School of Life Sciences, Shandong University, 27, Shanda Nan Road, Jinan 250100, China
| | - De-Li Shi
- School of Life Sciences, Shandong University, 27, Shanda Nan Road, Jinan 250100, China; Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, IBPS-Developmental Biology Laboratory, 75005 Paris, France.
| |
Collapse
|
6
|
Kjolby RAS, Harland RM. Genome-wide identification of Wnt/β-catenin transcriptional targets during Xenopus gastrulation. Dev Biol 2017; 426:165-175. [PMID: 27091726 PMCID: PMC6288011 DOI: 10.1016/j.ydbio.2016.03.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 10/21/2022]
Abstract
The canonical Wnt/β-catenin signaling pathway plays multiple roles during Xenopus gastrulation, including posteriorization of the neural plate, patterning of the mesoderm, and induction of the neural crest. Wnt signaling stabilizes β-catenin, which then activates target genes. However, few targets of this signaling pathway that mediate early developmental processes are known. Here we sought to identify transcriptional targets of the Wnt/β-catenin signaling pathway using a genome-wide approach. We selected putative targets using the criteria of reduced expression upon zygotic Wnt knockdown, β-catenin binding within 50kb of the gene, and expression in tissues that receive Wnt signaling. Using these criteria, we found 21 novel direct transcriptional targets of Wnt/β-catenin signaling during gastrulation and in addition have identified putative regulatory elements for further characterization in future studies.
Collapse
Affiliation(s)
- Rachel A S Kjolby
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Richard M Harland
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
7
|
Liu Y, Conboy I. Unexpected evolutionarily conserved rapid effects of viral infection on oxytocin receptor and TGF-β/pSmad3. Skelet Muscle 2017; 7:7. [PMID: 28506310 PMCID: PMC5433165 DOI: 10.1186/s13395-017-0125-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022] Open
Abstract
Background shRNA lentiviral vectors are extensively used for gene knockdowns in mammalian cells, and non-target shRNAs typically are considered the proper experimental control for general changes caused by RNAi. However, the effects of non-target lentivirus controls on the modulation of cell signaling pathways remain largely unknown. In this study, we evaluated the effect of control lentiviral transduction on oxytocin receptor (OXTR) expression through the ERK/MAPK pathway in mouse and human skeletal muscle cells, on myogenic activity, and in vivo on mouse muscle regeneration. Furthermore, we mined published data for the influence of viral infections on OXTR levels in human populations and found that unrelated viral pathologies have a common consequence: diminished levels of OXTR. Methods We examined the change in OXTR mRNA expression upon transduction with control and Smad3-targeting viral vectors through real time RT-PCR and Western blotting, and confirmed with immunofluorescence. Changes in Smad3 and OXTR expression were examined both in vitro with mouse and human myoblasts and in vivo in mouse satellite cells. The general effects of viral infections on OXTR downregulation in humans were also examined by analyzing published Gene Expression Omnibus (GEO) datasets. The change in myoblast myogenic activity caused by the viral transduction (the percent of Pax7 + Ki67+ cells) was examined by immunofluorescence. Results Results shown in this work establish that lentiviral control vectors significantly downregulate OXTR expression at mRNA and protein levels and diminish key downstream effectors of OXTR, ERK signaling, reducing the myogenic proliferation of infected cells. This effect is evolutionarily conserved between mouse and human myogenic cells, and it manifests in satellite cells after control lentiviral transduction of mice in vivo. Furthermore, an examination of published datasets uncovered similar OXTR downregulation in humans that are afflicted with different viral infections. Additionally, cells transduced with Smad3-targeting shRNA downregulate OXTR even more than cells transduced with control viruses. Conclusions Our work suggests that experimental cohorts transduced with control viruses may not behave the same as un-transduced cells and animals, specifically that control viral vectors significantly change the intensity of key cell-signaling pathways, such as OXTR/ERK. Our results further demonstrate that lentiviral transduction significantly decreases myogenic proliferation and suggest that viral infections in general may play a role in decreasing muscle health and regeneration, a decline in metabolic health, and a lower sense of well-being, as these rely on effective OXTR signaling. Additionally, our data suggest pathway crosstalk between TGF-β/pSmad3 and OXTR, implying that sustained attenuation of the TGF-β/pSmad3 pathway will reduce pro-regenerative OXTR/pERK signaling. Electronic supplementary material The online version of this article (doi:10.1186/s13395-017-0125-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Bioengineering and QB3 Institute, Univerisity of California, Berkeley, 174 Stanley Hall, Berkeley, CA, 94720, USA.
| | - Irina Conboy
- Department of Bioengineering and QB3 Institute, Univerisity of California, Berkeley, 174 Stanley Hall, Berkeley, CA, 94720, USA.
| |
Collapse
|
8
|
Genovese NJ, Domeier TL, Telugu BPVL, Roberts RM. Enhanced Development of Skeletal Myotubes from Porcine Induced Pluripotent Stem Cells. Sci Rep 2017; 7:41833. [PMID: 28165492 PMCID: PMC5292944 DOI: 10.1038/srep41833] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/30/2016] [Indexed: 02/07/2023] Open
Abstract
The pig is recognized as a valuable model in biomedical research in addition to its agricultural importance. Here we describe a means for generating skeletal muscle efficiently from porcine induced pluripotent stem cells (piPSC) in vitro thereby providing a versatile platform for applications ranging from regenerative biology to the ex vivo cultivation of meat. The GSK3B inhibitor, CHIR99021 was employed to suppress apoptosis, elicit WNT signaling events and drive naïve-type piPSC along the mesoderm lineage, and, in combination with the DNA methylation inhibitor 5-aza-cytidine, to activate an early skeletal muscle transcription program. Terminal differentiation was then induced by activation of an ectopically expressed MYOD1. Myotubes, characterized by myofibril development and both spontaneous and stimuli-elicited excitation-contraction coupling cycles appeared within 11 days. Efficient lineage-specific differentiation was confirmed by uniform NCAM1 and myosin heavy chain expression. These results provide an approach for generating skeletal muscle that is potentially applicable to other pluripotent cell lines and to generating other forms of muscle.
Collapse
Affiliation(s)
- Nicholas J Genovese
- C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 6521, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Bhanu Prakash V L Telugu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA ARS, Beltsville, MD 20705, USA
| | - R Michael Roberts
- C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 6521, USA
| |
Collapse
|
9
|
Sabillo A, Ramirez J, Domingo CR. Making muscle: Morphogenetic movements and molecular mechanisms of myogenesis in Xenopus laevis. Semin Cell Dev Biol 2016; 51:80-91. [PMID: 26853935 DOI: 10.1016/j.semcdb.2016.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
Abstract
Xenopus laevis offers unprecedented access to the intricacies of muscle development. The large, robust embryos make it ideal for manipulations at both the tissue and molecular level. In particular, this model system can be used to fate map early muscle progenitors, visualize cell behaviors associated with somitogenesis, and examine the role of signaling pathways that underlie induction, specification, and differentiation of muscle. Several characteristics that are unique to X. laevis include myogenic waves with distinct gene expression profiles and the late formation of dermomyotome and sclerotome. Furthermore, myogenesis in the metamorphosing frog is biphasic, facilitating regeneration studies. In this review, we describe the morphogenetic movements that shape the somites and discuss signaling and transcriptional regulation during muscle development and regeneration. With recent advances in gene editing tools, X. laevis remains a premier model organism for dissecting the complex mechanisms underlying the specification, cell behaviors, and formation of the musculature system.
Collapse
Affiliation(s)
- Armbien Sabillo
- Department of Molecular & Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Julio Ramirez
- Department of Biology, San Francisco State University, CA 94132, USA
| | - Carmen R Domingo
- Department of Biology, San Francisco State University, CA 94132, USA.
| |
Collapse
|
10
|
Li HY, Grifone R, Saquet A, Carron C, Shi DL. The Xenopus homologue of Down syndrome critical region protein 6 drives dorsoanterior gene expression and embryonic axis formation by antagonising polycomb group proteins. Development 2013; 140:4903-13. [DOI: 10.1242/dev.098319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mesoderm and embryonic axis formation in vertebrates is mediated by maternal and zygotic factors that activate the expression of target genes. Transcriptional derepression plays an important role in the regulation of expression in different contexts; however, its involvement and possible mechanism in mesoderm and embryonic axis formation are largely unknown. Here we demonstrate that XDSCR6, a Xenopus homologue of human Down syndrome critical region protein 6 (DSCR6, or RIPPLY3), regulates mesoderm and embryonic axis formation through derepression of polycomb group (PcG) proteins. Xdscr6 maternal mRNA is enriched in the endoderm of the early gastrula and potently triggers the formation of dorsal mesoderm and neural tissues in ectoderm explants; it also dorsalises ventral mesoderm during gastrulation and induces a secondary embryonic axis. A WRPW motif, which is present in all DSCR6 homologues, is necessary and sufficient for the dorsal mesoderm- and axis-inducing activity. Knockdown of Xdscr6 inhibits dorsal mesoderm gene expression and results in head deficiency. We further show that XDSCR6 physically interacts with PcG proteins through the WRPW motif, preventing the formation of PcG bodies and antagonising their repressor activity in embryonic axis formation. By chromatin immunoprecipitation, we demonstrate that XDSCR6 releases PcG proteins from chromatin and allows dorsal mesoderm gene transcription. Our studies suggest that XDSCR6 might function to sequester PcG proteins and identify a novel derepression mechanism implicated in embryonic induction and axis formation.
Collapse
Affiliation(s)
- Hong-Yan Li
- Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
- Department of Marine Biology, Ocean University of China, Qingdao 266003, China
| | - Raphaëlle Grifone
- Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
| | - Audrey Saquet
- Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
| | - Clémence Carron
- Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
| | - De-Li Shi
- Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
| |
Collapse
|
11
|
Wong J, Mehta V, Voronova A, Coutu J, Ryan T, Shelton M, Skerjanc IS. β-catenin is essential for efficient in vitro premyogenic mesoderm formation but can be partially compensated by retinoic acid signalling. PLoS One 2013; 8:e57501. [PMID: 23460868 PMCID: PMC3583846 DOI: 10.1371/journal.pone.0057501] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 01/24/2013] [Indexed: 12/14/2022] Open
Abstract
Previous studies have shown that P19 cells expressing a dominant negative β-catenin mutant (β-cat/EnR) cannot undergo myogenic differentiation in the presence or absence of muscle-inducing levels of retinoic acid (RA). While RA could upregulate premyogenic mesoderm expression, including Pax3/7 and Meox1, only Pax3/7 and Gli2 could be upregulated by RA in the presence of β-cat/EnR. However, the use of a dominant negative construct that cannot be compensated by other factors is limiting due to the possibility of negative chromatin remodelling overriding compensatory mechanisms. In this study, we set out to determine if β-catenin function is essential for myogenesis with and without RA, by creating P19 cells with reduced β-catenin transcriptional activity using an shRNA approach, termed P19[shβ-cat] cells. The loss of β-catenin resulted in a reduction of skeletal myogenesis in the absence of RA as early as premyogenic mesoderm, with the loss of Pax3/7, Eya2, Six1, Meox1, Gli2, Foxc1/2, and Sox7 transcript levels. Chromatin immunoprecipitation identified an association of β-catenin with the promoter region of the Sox7 gene. Differentiation of P19[shβ-cat] cells in the presence of RA resulted in the upregulation or lack of repression of all of the precursor genes, on day 5 and/or 9, with the exception of Foxc2. However, expression of Sox7, Gli2, the myogenic regulatory factors and terminal differentiation markers remained inhibited on day 9 and overall skeletal myogenesis was reduced. Thus, β-catenin is essential for in vitro formation of premyogenic mesoderm, leading to skeletal myogenesis. RA can at least partially compensate for the loss of β-catenin in the expression of many myogenic precursor genes, but not for myoblast gene expression or overall myogenesis.
Collapse
Affiliation(s)
- Jacob Wong
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Virja Mehta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Anastassia Voronova
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Josée Coutu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Tammy Ryan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael Shelton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ilona S. Skerjanc
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
12
|
Kong W, Yang Y, Zhang T, Shi DL, Zhang Y. Characterization of sFRP2-like in amphioxus: insights into the evolutionary conservation of Wnt antagonizing function. Evol Dev 2013; 14:168-77. [PMID: 23017025 DOI: 10.1111/j.1525-142x.2012.00533.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Wnt signaling plays a key role in embryonic patterning and morphogenetic movements. The secreted Frizzled-related proteins (sFRPs) antagonize Wnt signaling, but their roles in development are poorly understood. To determine whether function of sFRPs is conserved between amphioxus and vertebrates, we characterized sFRP2-like function in the amphioxus, Branchiostoma belcheri tsingtauense (B. belcheri). As in other species of Branchiostome, in B. belcheri, expression of sFRP2-like is restricted to the mesendoderm during gastrulation and to the anterior mesoderm and endoderm during neurulation. Functional analyses in frog (Xenopus laevis) indicate that amphioxus sFRP2-like potently inhibits both canonical and non-canonical Wnts. Thus, sFRP-2 probably functions in amphioxus embryos to inhibit Wnt signaling anteriorly. Moreover, dorsal overexpression of amphioxus sFRP2-like in Xenopus embryos, like inhibition of Wnt11, blocks gastrulation movements. This implies that sFRP2-like may also modulate Wnt signaling during gastrulation movements in amphioxus.
Collapse
Affiliation(s)
- Weihua Kong
- Institute of Developmental Biology, School of Life Sciences, Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Shandong University, Jinan, 250100, China
| | | | | | | | | |
Collapse
|
13
|
Key signalling factors and pathways in the molecular determination of skeletal muscle phenotype. Animal 2012; 1:681-98. [PMID: 22444469 DOI: 10.1017/s1751731107702070] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The molecular basis and control of the biochemical and biophysical properties of skeletal muscle, regarded as muscle phenotype, are examined in terms of fibre number, fibre size and fibre types. A host of external factors or stimuli, such as ligand binding and contractile activity, are transduced in muscle into signalling pathways that lead to protein modifications and changes in gene expression which ultimately result in the establishment of the specified phenotype. In skeletal muscle, the key signalling cascades include the Ras-extracellular signal regulated kinase-mitogen activated protein kinase (Erk-MAPK), the phosphatidylinositol 3'-kinase (PI3K)-Akt1, p38 MAPK, and calcineurin pathways. The molecular effects of external factors on these pathways revealed complex interactions and functional overlap. A major challenge in the manipulation of muscle of farm animals lies in the identification of regulatory and target genes that could effect defined and desirable changes in muscle quality and quantity. To this end, recent advances in functional genomics that involve the use of micro-array technology and proteomics are increasingly breaking new ground in furthering our understanding of the molecular determinants of muscle phenotype.
Collapse
|
14
|
Cao JM, Li SQ, Zhang HW, Shi DL. High mobility group B proteins regulate mesoderm formation and dorsoventral patterning during zebrafish and Xenopus early development. Mech Dev 2012; 129:263-74. [DOI: 10.1016/j.mod.2012.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
|
15
|
Li HY, Bourdelas A, Carron C, Shi DL. The RNA-binding protein Seb4/RBM24 is a direct target of MyoD and is required for myogenesis during Xenopus early development. Mech Dev 2010; 127:281-91. [PMID: 20338237 DOI: 10.1016/j.mod.2010.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 03/17/2010] [Accepted: 03/19/2010] [Indexed: 10/19/2022]
Abstract
RNA-binding proteins play an important role to post-transcriptionally regulate gene expression. During early development they exhibit temporally and spatially regulated expression pattern. The expression of Xenopus laevis Seb4 gene, also known as RBM24 in other vertebrates, is restricted to the lateral and ventral mesoderm during gastrulation and then localized to the somitic mesoderm, in a similar pattern as XMyoD gene. Using a hormone-inducible form of MyoD to identify potential direct MyoD target genes, we find that Seb4 expression is directly regulated by MyoD at the gastrula stage. We further show that a 0.65kb X. tropicalis RBM24 regulatory region contains multiple E boxes (CANNTG), which are potential binding sites for MyoD and other bHLH proteins. By injecting a RBM24 reporter construct into the animal pole of X. laevis embryos, we find that this reporter gene is indeed specifically activated by MyoD and repressed by a dominant negative MyoD mutant. Knockdown of Seb4 produces similar effects as those obtained by the dominant negative MyoD mutant, indicating that it is required for the expression of myogenic genes and myogenesis in the embryo. In cultured ectodermal explants, although overexpression of Seb4 has no obvious effect on myogenesis, knockdown of Seb4 inhibits the expression of myogenic genes and myogenesis induced by MyoD. These results reveal that Seb4 is a target of MyoD during myogenesis and is required for myogenic gene expression.
Collapse
Affiliation(s)
- Hong-Yan Li
- Groupe de Biologie Expérimentale, Laboratoire de Biologie du Développement, CNRS UMR 7622, Université Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France
| | | | | | | |
Collapse
|
16
|
Carlson ME, Conboy MJ, Hsu M, Barchas L, Jeong J, Agrawal A, Mikels AJ, Agrawal S, Schaffer DV, Conboy IM. Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 2009; 8:676-89. [PMID: 19732043 PMCID: PMC2783265 DOI: 10.1111/j.1474-9726.2009.00517.x] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Muscle stem (satellite) cells are relatively resistant to cell-autonomous aging. Instead, their endogenous signaling profile and regenerative capacity is strongly influenced by the aged P-Smad3, differentiated niche, and by the aged circulation. With respect to muscle fibers, we previously established that a shift from active Notch to excessive transforming growth factor-beta (TGF-β) induces CDK inhibitors in satellite cells, thereby interfering with productive myogenic responses. In contrast, the systemic inhibitor of muscle repair, elevated in old sera, was suggested to be Wnt. Here, we examined the age-dependent myogenic activity of sera TGF-β1, and its potential cross-talk with systemic Wnt. We found that sera TGF-β1 becomes elevated within aged humans and mice, while systemic Wnt remained undetectable in these species. Wnt also failed to inhibit satellite cell myogenicity, while TGF-β1 suppressed regenerative potential in a biphasic fashion. Intriguingly, young levels of TGF-β1 were inhibitory and young sera suppressed myogenesis if TGF-β1 was activated. Our data suggest that platelet-derived sera TGF-β1 levels, or endocrine TGF-β1 levels, do not explain the age-dependent inhibition of muscle regeneration by this cytokine. In vivo, TGF-β neutralizing antibody, or a soluble decoy, failed to reduce systemic TGF-β1 and rescue myogenesis in old mice. However, muscle regeneration was improved by the systemic delivery of a TGF-β receptor kinase inhibitor, which attenuated TGF-β signaling in skeletal muscle. Summarily, these findings argue against the endocrine path of a TGF-β1-dependent block on muscle regeneration, identify physiological modalities of age-imposed changes in TGF-β1, and introduce new therapeutic strategies for the broad restoration of aged organ repair.
Collapse
Affiliation(s)
- Morgan E Carlson
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA 94720, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Franchini A, Casarini L, Malagoli D, Ottaviani E. Expression of the genes siamois, engrailed-2, bmp4 and myf5 during Xenopus development in presence of the marine toxins okadaic acid and palytoxin. CHEMOSPHERE 2009; 77:308-312. [PMID: 19683326 DOI: 10.1016/j.chemosphere.2009.07.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/25/2009] [Accepted: 07/13/2009] [Indexed: 05/28/2023]
Abstract
The present investigation examines the effects of the marine toxins, okadaic acid (OA) and palytoxin (PTX), on some genes involved in the neural and muscular specification and patterning of Xenopus laevis. The RT-PCR analyses performed at different stages of embryonic and larval development (stages 11-47) demonstrated that both toxins induce an over-expression of the genes siamois and engrailed-2 and a different behaviour in bmp4 and myf5. Indeed, OA provoked a significant increase in bmp4 in the earliest stage (11) examined, a down-regulation from stages 12 to 17, and a renewed increase from the beginning of hatching onwards (stages 35-47). In contrast, myf5 was up-regulated in all stages up to 35. PTX induced an over-expression of both bmp4 and myf5 during the embryonic and early larval development stages. The results show that PTX induces an increase in expression levels in all tested genes, while the response to OA seems to be more stage-dependent, with the embryonic development stage more sensitive to the toxin than the larval stages.
Collapse
Affiliation(s)
- Antonella Franchini
- Department of Animal Biology, University of Modena and Reggio Emilia, via Campi 213/D, 41100 Modena, Italy
| | | | | | | |
Collapse
|
18
|
Garnett AT, Han TM, Gilchrist MJ, Smith JC, Eisen MB, Wardle FC, Amacher SL. Identification of direct T-box target genes in the developing zebrafish mesoderm. Development 2009; 136:749-60. [PMID: 19158186 PMCID: PMC2685943 DOI: 10.1242/dev.024703] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2008] [Indexed: 11/20/2022]
Abstract
The zebrafish genes spadetail (spt) and no tail (ntl) encode T-box transcription factors that are important for early mesoderm development. Although much has been done to characterize these genes, the identity and location of target regulatory elements remain largely unknown. Here, we survey the genome for downstream target genes of the Spt and Ntl T-box transcription factors. We find evidence for extensive additive interactions towards gene activation and limited evidence for combinatorial and antagonistic interactions between the two factors. Using in vitro binding selection assays to define Spt- and Ntl-binding motifs, we searched for target regulatory sequence via a combination of binding motif searches and comparative genomics. We identified regulatory elements for tbx6 and deltaD, and, using chromatin immunoprecipitation, in vitro DNA binding assays and transgenic methods, we provide evidence that both are directly regulated by T-box transcription factors. We also find that deltaD is directly activated by T-box factors in the tail bud, where it has been implicated in starting the segmentation clock, suggesting that spt and ntl act upstream of this process.
Collapse
Affiliation(s)
- Aaron T Garnett
- Department of Molecular and Cell Biology and Center for Integrative Genomics, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Barillot W, Tréguer K, Faucheux C, Fédou S, Thézé N, Thiébaud P. Induction and modulation of smooth muscle differentiation in Xenopus embryonic cells. Dev Dyn 2009; 237:3373-86. [PMID: 18855898 DOI: 10.1002/dvdy.21749] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
By comparison with skeletal or cardiac developmental programs, little is known regarding the specific factors that promote specification and differentiation of smooth muscle cells from pluripotent cells. We have analyzed the developmental expression of a subset of smooth muscle genes during Xenopus early development and showed that similar to mammals and avians, Xenopus smooth muscle myosin heavy chain (SM-MHC) is a highly specific marker of smooth muscle differentiation. Embryonic cells from animal pole explants of Xenopus blastula can be induced by basic fibroblast growth factor, Wnt, and bone morphogenetic protein signals to adopt the smooth muscle pathway. Explants from early embryos that contain neural crest cells can also differentiate into cells expressing smooth muscle genes. We examined the interplay of several transcription factors, that is SRF, myocardin, and GATA6, that induce the expression of SM-MHC in animal cap cells and found that myocardin-dependent expression of smooth muscle genes in animal cap cells is synergized by SRF but is strongly antagonized by GATA6.
Collapse
|
20
|
Keren A, Keren-Politansky A, Bengal E. A p38 MAPK-CREB pathway functions to pattern mesoderm in Xenopus. Dev Biol 2008; 322:86-94. [PMID: 18675264 DOI: 10.1016/j.ydbio.2008.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 06/29/2008] [Accepted: 07/07/2008] [Indexed: 12/30/2022]
Abstract
Dorsal-ventral patterning is specified by signaling centers secreting antagonizing morphogens that form a signaling gradient. Yet, how morphogen gradient is translated intracellularly into fate decisions remains largely unknown. Here, we report that p38 MAPK and CREB function along the dorsal-ventral axis in mesoderm patterning. We find that the phosphorylated form of CREB (S133) is distributed in a gradient along the dorsal-ventral mesoderm axis and that the p38 MAPK pathway mediates the phosphorylation of CREB. Knockdown of CREB prevents chordin expression and mesoderm dorsalization by the Spemann organizer, whereas ectopic expression of activated CREB-VP16 chimera induces chordin expression and dorsalizes mesoderm. Expression of high levels of p38 activator, MKK6E or CREB-VP16 in embryos converts ventral mesoderm into a dorsal organizing center. p38 MAPK and CREB function downstream of maternal Wnt/beta-catenin and the organizer-specific genes siamois and goosecoid. At low expression levels, MKK6E induces expression of lateral genes without inducing the expression of dorsal genes. Loss of CREB or p38 MAPK activity enables the expansion of the ventral homeobox gene vent1 into the dorsal marginal region, preventing the lateral expression of Xmyf5. Overall, these data indicate that dorsal-ventral mesoderm patterning is regulated by differential p38/CREB activities along the axis.
Collapse
Affiliation(s)
- Aviad Keren
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| | | | | |
Collapse
|
21
|
Abstract
Immunodetection of beta-catenin accumulation in the nucleus is the most direct and reliable method to determine the intensity and the spatial/temporal patterns of Wnt-dependent signaling activity. Due to the large size of the Xenopus embryo, staining must be done on sections. We present here a simple protocol to prepare cryosections and produce high-quality images of the early embryo using immunofluorescence. We also provide comments on various conceptual and technical issues from fixation to image collection, which may assist in optimizing immunodetection in embryos and tissues beyond the specific scope of beta-catenin localization.
Collapse
|
22
|
Multiple upstream modules regulate zebrafish myf5 expression. BMC DEVELOPMENTAL BIOLOGY 2007; 7:1. [PMID: 17199897 PMCID: PMC1769357 DOI: 10.1186/1471-213x-7-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Accepted: 01/03/2007] [Indexed: 11/27/2022]
Abstract
Background Myf5 is one member of the basic helix-loop-helix family of transcription factors, and it functions as a myogenic factor that is important for the specification and differentiation of muscle cells. The expression of myf5 is somite- and stage-dependent during embryogenesis through a delicate regulation. However, this complex regulatory mechanism of myf5 is not clearly understood. Results We isolated a 156-kb bacterial artificial chromosome clone that includes an upstream 80-kb region and a downstream 70-kb region of zebrafish myf5 and generated a transgenic line carrying this 156-kb segment fused to a green fluorescent protein (GFP) reporter gene. We find strong GFP expression in the most rostral somite and in the presomitic mesoderm during segmentation stages, similar to endogenous myf5 expression. Later, the GFP signals persist in caudal somites near the tail bud but are down-regulated in the older, rostral somites. During the pharyngula period, we detect GFP signals in pectoral fin buds, dorsal rostral myotomes, hypaxial myotomes, and inferior oblique and superior oblique muscles, a pattern that also corresponds well with endogenous myf5 transcripts. To characterize the specific upstream cis-elements that regulate this complex and dynamic expression pattern, we also generated several transgenic lines that harbor various lengths within the upstream 80-kb segment. We find that (1) the -80 kb/-9977 segment contains a fin and cranial muscle element and a notochord repressor; (2) the -9977/-6213 segment contains a strong repressive element that does not include the notochord-specific repressor; (3) the -6212/-2938 segment contains tissue-specific elements for bone and spinal cord; (4) the -2937/-291 segment contains an eye enhancer, and the -2937/-2457 segment is required for notochord and myocyte expression; and (5) the -290/-1 segment is responsible for basal transcription in somites and the presomitic mesoderm. Conclusion We suggest that the cell lineage-specific expression of myf5 is delicately orchestrated by multiple modules within the distal upstream region. This study provides an insight to understand the molecular control of myf5 and myogenesis in the zebrafish.
Collapse
|
23
|
Walston T, Guo C, Proenca R, Wu M, Herman M, Hardin J, Hedgecock E. mig-5/Dsh controls cell fate determination and cell migration in C. elegans. Dev Biol 2006; 298:485-97. [PMID: 16899238 DOI: 10.1016/j.ydbio.2006.06.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2006] [Revised: 06/24/2006] [Accepted: 06/30/2006] [Indexed: 01/01/2023]
Abstract
Cell fate determination and cell migration are two essential events in the development of an organism. We identify mig-5, a Dishevelled family member, as a gene that regulates several cell fate decisions and cell migrations that are important during C. elegans embryonic and larval development. In offspring from mig-5 mutants, cell migrations are defective during hypodermal morphogenesis, QL neuroblast migration, and the gonad arm migration led by the distal tip cells (DTCs). In addition to abnormal migration, DTC fate is affected, resulting in either an absent or an extra DTC. The cell fates of the anchor cell in hermaphrodites and the linker cells in the male gonad are also defective, often resulting in the cells adopting the fates of their sister lineage. Moreover, 2 degrees vulval precursor cells occasionally adopt the 3 degrees vulval cell fate, resulting in a deformed vulva, and the P12 hypodermal precursor often differentiates into a second P11 cell. These defects demonstrate that MIG-5 is essential in determining proper cell fate and cell migration throughout C. elegans development.
Collapse
Affiliation(s)
- Timothy Walston
- Laboratory of Genetics, University of Wisconsin-Madison, 1117 W. Johnson St., Madison, WI 53706, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Li HY, Bourdelas A, Carron C, Gomez C, Boucaut JC, Shi DL. FGF8, Wnt8 and Myf5 are target genes of Tbx6 during anteroposterior specification in Xenopus embryo. Dev Biol 2006; 290:470-81. [PMID: 16343478 DOI: 10.1016/j.ydbio.2005.11.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 10/17/2005] [Accepted: 11/14/2005] [Indexed: 11/21/2022]
Abstract
The T-box transcription factor Tbx6 is required for somite formation and loss-of-function or reduced activity of Tbx6 result in absence of posterior paraxial mesoderm or disorganized somites, but how it is involved in a regulatory hierarchy during Xenopus early development is not clear. We show here that Tbx6 is expressed in the lateral and ventral mesoderm of early gastrula, and it is necessary and sufficient to directly and indirectly regulate the expression of a subset of early mesodermal and endodermal genes. Ectopic expression of Tbx6 inhibits early neuroectodermal gene expression by strongly inducing the expression of posterior mesodermal genes, and expands the mesoderm territory at the expense of neuroectoderm. Conversely, overexpression of a dominant negative Tbx6 mutant in the ventral mesoderm inhibits the expression of several mesodermal genes and results in neural induction in a dose-dependent manner. Using a hormone-inducible form of Tbx6, we have identified FGF8, Xwnt8 and XMyf5 as immediate early responsive genes of Tbx6, and the induction of these genes by Tbx6 is independent of Xbra and VegT. These target genes act downstream and mediate the function of Tbx6 in anteroposterior specification. Our results therefore identify a regulatory cascade governed by Tbx6 in the specification of posterior mesoderm during Xenopus early development.
Collapse
Affiliation(s)
- Hong-Yan Li
- Groupe de Biologie Expérimentale, Laboratoire de Biologie du Développement, CNRS UMR 7622, Université Pierre et Marie-Curie, 9 quai Saint-Bernard, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
25
|
Hamade A, Deries M, Begemann G, Bally-Cuif L, Genêt C, Sabatier F, Bonnieu A, Cousin X. Retinoic acid activates myogenesis in vivo through Fgf8 signalling. Dev Biol 2006; 289:127-40. [PMID: 16316642 DOI: 10.1016/j.ydbio.2005.10.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 09/13/2005] [Accepted: 10/21/2005] [Indexed: 11/21/2022]
Abstract
Retinoic acid (RA) has been shown to regulate muscle differentiation in vitro. Here, we have investigated the role of RA signalling during embryonic myogenesis in zebrafish. We have altered RA signalling from gastrulation stages onwards by either inhibiting endogenous RA synthesis using an inhibitor of retinaldehyde dehydrogenases (DEAB) or by addition of exogenous RA. DEAB reduces expression of the myogenic markers myoD and myogenin in somites, whereas RA induces increased expression of these genes and strongly induces premature myoD expression in the presomitic mesoderm (psm). The expression dynamics of myf5 in presomitic and somitic mesoderm suggest that RA promotes muscle differentiation, a role supported by the fact that RA activates expression of fast myosin, while DEAB represses it. We identify Fgf8 as a major relay factor in RA-mediated activation of myogenesis. We show that fgf8 expression in somites and anterior psm is regulated by RA, and find that in the absence of Fgf8 signalling in the acerebellar mutant RA fails to promote myoD expression. We propose that, in the developing embryo, localised synthesis of RA by Raldh2 in the anterior psm and in somites activates fgf8 expression which in turn induces the expression of myogenic genes and fast muscle differentiation.
Collapse
Affiliation(s)
- Aline Hamade
- UMR866 Différenciation Cellulaire et Croissance, INRA, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Keren A, Bengal E, Frank D. p38 MAP kinase regulates the expression of XMyf5 and affects distinct myogenic programs during Xenopus development. Dev Biol 2005; 288:73-86. [PMID: 16248994 DOI: 10.1016/j.ydbio.2005.09.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Revised: 09/06/2005] [Accepted: 09/08/2005] [Indexed: 12/30/2022]
Abstract
The p38 MAPK signaling pathway is essential for skeletal muscle differentiation in tissue culture models. We demonstrate a novel role for p38 MAPK in myogenesis during early Xenopus laevis development. Interfering with p38 MAPK causes distinct defects in myogenesis. The initial expression of Myf5 is selectively blocked, while expression of MyoD is unaffected. Expression of a subset of muscle structural genes is reduced. Convergent extension movements are prevented and segmentation of the paraxial mesoderm is delayed, probably due to the failure of cells to withdraw from the cell cycle. Myotubes are properly formed; however, at later stages, they begin to degenerate, and the boundaries between somites disappear. Significant apoptotic cell death occurs in most parts of the somites. The ventral body wall muscle derived from migratory progenitor cells of the ventral somite region is poorly formed. Our data indicate that the developmental defects caused by p38alpha-knockdown were mediated by the loss of XMyf5 expression. Thus, this study identifies a specific intracellular pathway in which p38 MAPK and Myf5 proteins regulate a distinct myogenic program.
Collapse
Affiliation(s)
- Aviad Keren
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| | | | | |
Collapse
|
27
|
Carron C, Bourdelas A, Li HY, Boucaut JC, Shi DL. Antagonistic interaction between IGF and Wnt/JNK signaling in convergent extension in Xenopus embryo. Mech Dev 2005; 122:1234-47. [PMID: 16169711 DOI: 10.1016/j.mod.2005.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 06/17/2005] [Accepted: 06/17/2005] [Indexed: 10/25/2022]
Abstract
The homeobox gene Otx2 is expressed during gastrulation in the anterior domain of the vertebrate embryo and is involved in neural and head induction during Xenopus early development. It also prevents convergent extension movements in trunk and posterior mesoderm. Insulin-like growth factors (IGFs) were shown to have similar function. However, whether they interact and the mechanism by which they affect convergent extension remain unclear. We show that IGF pathway specifically induces the expression of Otx2 in the early gastrula and blocks convergent extension of neuroectoderm and mesoderm through the transcriptional activation of Otx2 gene. Otx2 represses the expression of Xbra and Xwnt-11, and the effects of IGF on gastrulation movements can be partially rescued by antisense Otx2 morpholino oligonucleotide. These indicate that IGF pathway interacts with Otx2 to restrict Xbra and Xwnt-11 expression in the trunk and posterior regions. Consistent with this, we show that inhibition of IGF signaling or Otx2 function induces Xbra and Xwnt11 expression and convergent extension in ectodermal cells. Furthermore, the blockade of convergent extension by IGF-I and Otx2 can be rescued by coexpression of Xwnt-11 or a constitutively active Jun N-terminal kinase (JNK). Because Xbra and Xwnt-11 are required for convergent extension movements and Xwnt-11 activates the non-canonical Wnt-11/JNK pathway, our results reveal a mutually exclusive function between IGF and Wnt-11/JNK pathways in regulating cell behaviours during vertebrate head and trunk development.
Collapse
Affiliation(s)
- Clémence Carron
- Groupe de Biologie Expérimentale, Laboratoire de Biologie du Développement, CNRS UMR 7622, Université Paris 6, 9 quai Saint-Bernard, 75005 Paris, France
| | | | | | | | | |
Collapse
|
28
|
Kazanskaya O, Glinka A, del Barco Barrantes I, Stannek P, Niehrs C, Wu W. R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Xenopus myogenesis. Dev Cell 2004; 7:525-34. [PMID: 15469841 DOI: 10.1016/j.devcel.2004.07.019] [Citation(s) in RCA: 351] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 07/29/2004] [Accepted: 07/30/2004] [Indexed: 01/26/2023]
Abstract
We have carried out a small pool expression screen for modulators of the Wnt/beta-catenin pathway and identified Xenopus R-spondin2 (Rspo2) as a secreted activator of this cascade. Rspo2 is coexpressed with and positively regulated by Wnt signals and synergizes with Wnts to activate beta-catenin. Analyses of functional interaction with components of the Wnt/beta-catenin pathway suggest that Rspo2 functions extracellularly at the level of receptor ligand interaction. In addition to activating the Wnt/beta-catenin pathway, Rspo2 overexpression blocks Activin, Nodal, and BMP4 signaling in Xenopus, raising the possibility that it may negatively regulate the TGF-beta pathway. Antisense Morpholino experiments in Xenopus embryos and RNAi experiments in HeLa cells reveal that Rspo2 is required for Wnt/beta-catenin signaling. In Xenopus embryos depleted of Rspo2, the muscle markers myoD and myf5 fail to be activated and later muscle development is impaired. Thus, Rspo2 functions in a positive feedback loop to stimulate the Wnt/beta-catenin cascade.
Collapse
Affiliation(s)
- Olga Kazanskaya
- Division of Molecular Embryology, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Bourdelas A, Li HY, Boucaut JC, Shi DL. Identification of distinct genes with restricted expression in the somitic mesoderm in Xenopus embryo. Gene Expr Patterns 2004; 4:695-9. [PMID: 15465491 DOI: 10.1016/j.modgep.2004.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 04/05/2004] [Accepted: 04/06/2004] [Indexed: 10/26/2022]
Abstract
We have used whole-mount in situ hybridisation to identify genes expressed in the somitic mesoderm during Xenopus early development. We report here the analysis of eight genes whose expression pattern has not been described previously. They include the Xenopus homologues of eukaryotic initiation factor 2beta, methionine adenosyltransferase II, serine dehydratase, alpha-adducin, oxoglutarate dehydrogenase, fragile X mental retardation syndrome related protein 1, monocarboxylate transporter and voltage-dependent anion channel 1. Interestingly, these genes exhibit very dynamic expression pattern during early development. At early gastrula stages several genes do not show localised expression pattern, while other genes are expressed in the marginal mesoderm or in ectoderm. As development proceeds, the expression of these genes is gradually restricted to different compartments of somite. This study thus reveals an unexpected dynamic expression pattern for various genes with distinct function in vertebrates.
Collapse
Affiliation(s)
- Audrey Bourdelas
- Groupe de Biologie Expérimentale, Laboratoire de Biologie du Développement, CNRS UMR 7622, Université Paris VI, 9 quai Saint-Bernard, Paris 75005, France
| | | | | | | |
Collapse
|
30
|
de Boer J, Siddappa R, Gaspar C, van Apeldoorn A, Fodde R, van Blitterswijk C. Wnt signaling inhibits osteogenic differentiation of human mesenchymal stem cells. Bone 2004; 34:818-26. [PMID: 15121013 DOI: 10.1016/j.bone.2004.01.016] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Revised: 12/24/2003] [Accepted: 01/22/2004] [Indexed: 11/29/2022]
Abstract
Human mesenchymal stem cells (hMSCs) from the bone marrow represent a potential source of pluripotent cells for autologous bone tissue engineering. We previously discovered that over activation of the Wnt signal transduction pathway by either lithium or Wnt3A stimulates hMSC proliferation while retaining pluripotency. Release of Wnt3A or lithium from porous calcium phosphate scaffolds, which we use for bone tissue engineering, could provide a mitogenic stimulus to implanted hMSCs. To define the proper release profile, we first assessed the effect of Wnt over activation on osteogenic differentiation of hMSCs. Here, we report that both lithium and Wnt3A strongly inhibit dexamethasone-induced expression of the osteogenic marker alkaline phosphatase (ALP). Moreover, lithium partly inhibited mineralization of hMSCs whereas Wnt3A completely blocked it. Time course analysis during osteogenic differentiation revealed that 4 days of Wnt3A exposure before the onset of mineralization is sufficient to block mineralization completely. Gene expression profiling in Wnt3A and lithium-exposed hMSCs showed that many osteogenic and chondrogenic markers, normally expressed in proliferating hMSCs, are downregulated upon Wnt stimulation. We conclude that Wnt signaling inhibits dexamethasone-induced osteogenesis in hMSCs. In future studies, we will try to limit release of lithium or Wnt3A from calcium phosphate scaffolds to the proliferative phase of osteogenesis.
Collapse
Affiliation(s)
- Jan de Boer
- Institute for Biomedical Technology, University of Twente, Enschede, The Netherlands.
| | | | | | | | | | | |
Collapse
|
31
|
Pownall ME, Gustafsson MK, Emerson CP. Myogenic regulatory factors and the specification of muscle progenitors in vertebrate embryos. Annu Rev Cell Dev Biol 2003; 18:747-83. [PMID: 12142270 DOI: 10.1146/annurev.cellbio.18.012502.105758] [Citation(s) in RCA: 428] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Embryological and genetic studies of mouse, bird, zebrafish, and frog embryos are providing new insights into the regulatory functions of the myogenic regulatory factors, MyoD, Myf5, Myogenin, and MRF4, and the transcriptional and signaling mechanisms that control their expression during the specification and differentiation of muscle progenitors. Myf5 and MyoD genes have genetically redundant, but developmentally distinct regulatory functions in the specification and the differentiation of somite and head muscle progenitor lineages. Myogenin and MRF4 have later functions in muscle differentiation, and Pax and Hox genes coordinate the migration and specification of somite progenitors at sites of hypaxial and limb muscle formation in the embryo body. Transcription enhancers that control Myf5 and MyoD activation in muscle progenitors and maintain their expression during muscle differentiation have been identified by transgenic analysis. In epaxial, hypaxial, limb, and head muscle progenitors, Myf5 is controlled by lineage-specific transcription enhancers, providing evidence that multiple mechanisms control progenitor specification at different sites of myogenesis in the embryo. Developmental signaling ligands and their signal transduction effectors function both interactively and independently to control Myf5 and MyoD activation in muscle progenitor lineages, likely through direct regulation of their transcription enhancers. Future investigations of the signaling and transcriptional mechanisms that control Myf5 and MyoD in the muscle progenitor lineages of different vertebrate embryos can be expected to provide a detailed understanding of the developmental and evolutionary mechanisms for anatomical muscles formation in vertebrates. This knowledge will be a foundation for development of stem cell therapies to repair diseased and damaged muscles.
Collapse
|
32
|
Bain G, Müller T, Wang X, Papkoff J. Activated beta-catenin induces osteoblast differentiation of C3H10T1/2 cells and participates in BMP2 mediated signal transduction. Biochem Biophys Res Commun 2003; 301:84-91. [PMID: 12535644 DOI: 10.1016/s0006-291x(02)02951-0] [Citation(s) in RCA: 215] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Wnt glycoproteins are important regulators of cellular differentiation and embryonic development. Some Wnt proteins induce stabilization of beta-catenin which cooperatively regulates gene expression with LEF/Tcf transcription factors. Here we demonstrate a direct role for beta-catenin signaling in osteoblast differentiation and in BMP2-mediated signal transduction. Similar to treatment with BMP-2 protein, ectopic expression of stabilized beta-catenin in C3H10T1/2 cells or activation of endogenous beta-catenin signaling with LiCl induces expression of alkaline phosphatase mRNA and protein, a defined marker of early osteoblast differentiation. Unlike BMP2 protein, stabilized beta-catenin does not induce osteocalcin gene expression, a marker of late osteoblast differentiation. BMP2-induced differentiation also leads to activation of endogenous beta-catenin signaling thus implicating beta-catenin in early steps of BMP2-mediated osteoblast differentiation. Effects of beta-catenin and BMP2 on C3H10T1/2 differentiation are not completely overlapping, implying that some aspects of BMP2-induced differentiation may be mediated by beta-catenin signaling and that beta-catenin can also participate in non-BMP2-dependent differentiation processes.
Collapse
Affiliation(s)
- Gerard Bain
- Aventis Cambridge Genomics Center, 26 Landsdowne Street, Cambridge, MA 02139, USA.
| | | | | | | |
Collapse
|
33
|
Abstract
Xenopus myogenesis is characterized by specific features, different from those of mammalian and avian systems both at the cellular level and in gene expression patterns. During early embryogenesis, after the initial molecular signals inducing mesoderm, the myogenic determination factors XMyoD and XMyf-5 are activated in presomitic mesoderm in response to mesoderm-inducing factors. After these first inductions of the myogenic program, forming muscles in Xenopus can have different destinies, some of these resulting in cell death before adulthood. In particular, it is quite characteristic of this species that, during metamorphosis, the primary myotomal myofibers completely die and are progressively replaced by secondary "adult" multinucleated myofibers. This feature offers the unique opportunity to totally separate the molecular analysis of these two distinct types of myogenesis. The aim of this review is to summarize our knowledge on the cellular and molecular events as well as the epigenetic regulations involved in the construction of Xenopus muscles during development.
Collapse
Affiliation(s)
- Christophe Chanoine
- Laboratoire de Biologie du Développement et de la Différenciation Musculaire (LNRS UMR 7060 CNRS), Paris, France.
| | | |
Collapse
|
34
|
Lin GF, Geng X, Chen Y, Qu B, Wang F, Hu R, Ding X. T-box binding site mediates the dorsal activation of myf-5 in Xenopus gastrula embryos. Dev Dyn 2003; 226:51-8. [PMID: 12508224 DOI: 10.1002/dvdy.10215] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Myf-5, a member of the muscle regulatory factor family of transcription factors, plays an important role in the determination, development, and differentiation of the skeletal muscle. Factors that regulate the expression of myf-5 itself are not well understood. We show here that a T-box binding site in the Xenopus myf-5 promoter mediated the activation of myf-5 expression through specific interaction with nuclear proteins of gastrula embryos. The T-box binding site could be bound by and respond to T-box proteins. T-box genes could induce Xmyf-5. The results suggest that T-box proteins are involved in the specification of myogenic mesoderm and muscle development.
Collapse
Affiliation(s)
- Gu Fa Lin
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|