1
|
Miao D, Ren J, Jia Y, Jia Y, Li Y, Huang H, Gao R. PAX1 represses canonical Wnt signaling pathway and plays dual roles during endoderm differentiation. Cell Commun Signal 2024; 22:242. [PMID: 38664733 PMCID: PMC11046865 DOI: 10.1186/s12964-024-01629-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Paired box 1 (PAX1) is a transcription factor and essential for the development of pharyngeal pouches-derived tissues, including thymus. PAX1 mutations are identified in Severe Combined Immunodeficiency (SCID) patients with Otofaciocervical Syndrome Type 2 (OTFCS2). However, despite the critical roles of PAX1 in embryonic development and diseases, detailed insights into its molecular mode of action are critically missing. METHODS The repressing roles of PAX1 and SCID associated mutants on Wnt signaling pathway were investigated by luciferase reporter assays, qRT-PCR and in situ hybridization in HEK293FT, HCT116 cells and zebrafish embryos, respectively. Co-immunoprecipitation (co-IP) and western blotting assays were carried out to identify the molecular mechanisms underlying PAX1's role on Wnt signaling pathway. hESC based endoderm differentiation, flow cytometry, high-throughput sequencing data analysis, and qRT-PCR assays were utilized to determine the roles of PAX1 during endoderm differentiation. RESULTS Here, we show that PAX1 represses canonical Wnt signaling pathway in vertebrate cells. Mechanically, PAX1 competes with SUMO E3 ligase PIASy to bind to TCF7L2, thus perturbing TCF7L2 SUMOylation level, further reducing its transcriptional activity and protein stability. Moreover, we reveal that PAX1 plays dual roles in hESC-derived definitive and foregut/pharyngeal endoderm cells, which give rise to the thymus epithelium, by inhibiting Wnt signaling. Importantly, our data show PAX1 mutations found in SCID patients significantly compromise the suppressing ability of PAX1 on Wnt signaling. CONCLUSIONS Our study presents a novel molecular mode of action of PAX1 in regulation of canonical Wnt signaling and endoderm differentiation, thus providing insights for the molecular basis of PAX1 associated SCID, offering better understanding of the behavior of PAX1 in embryogenesis.
Collapse
Affiliation(s)
- Danxiu Miao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, 150000, China
| | - Jie Ren
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
| | - Yanhan Jia
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Yihui Jia
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
| | - Yanshu Li
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, 150000, China
- College of Public Health, Shantou University, Shantou, 515063, China
| | - Huizhe Huang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rui Gao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
2
|
Li G, Strong A, Wang H, Kim JS, Watson D, Zhao S, Vaccaro C, Hartung E, Hakonarson H, Zhang TJ, Giampietro PF, Wu N. TBX6 as a cause of a combined skeletal-kidney dysplasia syndrome. Am J Med Genet A 2022; 188:3469-3481. [PMID: 36161696 PMCID: PMC10473889 DOI: 10.1002/ajmg.a.62972] [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: 04/12/2022] [Revised: 06/24/2022] [Accepted: 08/06/2022] [Indexed: 01/31/2023]
Abstract
TBX6 encodes transcription-factor box 6, a transcription factor critical to paraxial mesoderm segmentation and somitogenesis during embryonic development. TBX6 haploinsufficiency is believed to drive the skeletal and kidney phenotypes associated with the 16p11.2 deletion syndrome. Heterozygous and biallelic variants in TBX6 are associated with vertebral and rib malformations (TBX6-associated congenital scoliosis) and spondylocostal dysostosis, and heterozygous TBX6 variants are associated with increased risk of genitourinary tract malformations. Combined skeletal and kidney phenotypes in individuals harboring heterozygous or biallelic TBX6 variants are rare. Here, we present seven individuals with vertebral and rib malformations and structural kidney differences associated with heterozygous TBX6 gene deletion in trans with a hypomorphic TBX6 allele or biallelic TBX6 variants. Our case series highlights the association between TBX6 and both skeletal and kidney disease.
Collapse
Affiliation(s)
- Guozhuang Li
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Alanna Strong
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Haojun Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ji-Sun Kim
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical Center, New Brunswick, NJ
| | - Deborah Watson
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Courtney Vaccaro
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Erum Hartung
- Division of Nephrology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hakon Hakonarson
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Philip F. Giampietro
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical Center, New Brunswick, NJ
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL
| | - Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| |
Collapse
|
3
|
Martin BL. Mesoderm induction and patterning: Insights from neuromesodermal progenitors. Semin Cell Dev Biol 2022; 127:37-45. [PMID: 34840081 PMCID: PMC9130346 DOI: 10.1016/j.semcdb.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/23/2022]
Abstract
The discovery of mesoderm inducing signals helped usher in the era of molecular developmental biology, and today the mechanisms of mesoderm induction and patterning are still intensely studied. Mesoderm induction begins during gastrulation, but recent evidence in vertebrates shows that this process continues after gastrulation in a group of posteriorly localized cells called neuromesodermal progenitors (NMPs). NMPs reside within the post-gastrulation embryonic structure called the tailbud, where they make a lineage decision between ectoderm (spinal cord) and mesoderm. The majority of NMP-derived mesoderm generates somites, but also contributes to lateral mesoderm fates such as endothelium. The discovery of NMPs provides a new paradigm in which to study vertebrate mesoderm induction. This review will discuss mechanisms of mesoderm induction within NMPs, and how they have informed our understanding of mesoderm induction more broadly within vertebrates as well as animal species outside of the vertebrate lineage. Special focus will be given to the signaling networks underlying NMP-derived mesoderm induction and patterning, as well as emerging work on the significance of partial epithelial-mesenchymal states in coordinating cell fate and morphogenesis.
Collapse
Affiliation(s)
- Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
4
|
Cang Z, Nie Q. Inferring spatial and signaling relationships between cells from single cell transcriptomic data. Nat Commun 2020; 11:2084. [PMID: 32350282 PMCID: PMC7190659 DOI: 10.1038/s41467-020-15968-5] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/27/2020] [Indexed: 01/20/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) provides details for individual cells; however, crucial spatial information is often lost. We present SpaOTsc, a method relying on structured optimal transport to recover spatial properties of scRNA-seq data by utilizing spatial measurements of a relatively small number of genes. A spatial metric for individual cells in scRNA-seq data is first established based on a map connecting it with the spatial measurements. The cell-cell communications are then obtained by "optimally transporting" signal senders to target signal receivers in space. Using partial information decomposition, we next compute the intercellular gene-gene information flow to estimate the spatial regulations between genes across cells. Four datasets are employed for cross-validation of spatial gene expression prediction and comparison to known cell-cell communications. SpaOTsc has broader applications, both in integrating non-spatial single-cell measurements with spatial data, and directly in spatial single-cell transcriptomics data to reconstruct spatial cellular dynamics in tissues.
Collapse
Affiliation(s)
- Zixuan Cang
- Department of Mathematics, University of California, Irvine, Irvine, CA, 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, 92697, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
5
|
Große A, Perner B, Naumann U, Englert C. Zebrafish Wtx is a negative regulator of Wnt signaling but is dispensable for embryonic development and organ homeostasis. Dev Dyn 2019; 248:866-881. [PMID: 31290212 DOI: 10.1002/dvdy.84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The X-chromosomally linked gene WTX is a human disease gene and a member of the AMER family. Mutations in WTX are found in Wilms tumor, a form of pediatric kidney cancer and in patients suffering from OSCS (Osteopathia striata with cranial sclerosis), a sclerosing bone disorder. Functional data suggest WTX to be an inhibitor of the Wnt/β-catenin signaling pathway. Deletion of Wtx in mouse leads to perinatal death, impeding the analysis of its physiological role. RESULTS To gain insights into the function of Wtx in development and homeostasis we have used zebrafish as a model and performed both knockdown and knockout studies using morpholinos and transcription activator-like effector nucleases (TALENs), respectively. Wtx knockdown led to increased Wnt activity and embryonic dorsalization. Also, wtx mutants showed a transient upregulation of Wnt target genes in the context of caudal fin regeneration. Surprisingly, however, wtx as well as wtx/amer2/amer3 triple mutants developed normally, were fertile and did not show any anomalies in organ maintenance. CONCLUSIONS Our data show that members of the zebrafish wtx/amer gene family, while sharing a partially overlapping expression pattern do not compensate for each other. This observation demonstrates a remarkable robustness during development and regeneration in zebrafish.
Collapse
Affiliation(s)
- Andreas Große
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Birgit Perner
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Uta Naumann
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Christoph Englert
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
6
|
Organization of Embryonic Morphogenesis via Mechanical Information. Dev Cell 2019; 49:829-839.e5. [PMID: 31178400 DOI: 10.1016/j.devcel.2019.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 03/20/2019] [Accepted: 05/03/2019] [Indexed: 01/19/2023]
Abstract
Embryonic organizers establish gradients of diffusible signaling molecules to pattern the surrounding cells. Here, we elucidate an additional mechanism of embryonic organizers that is a secondary consequence of morphogen signaling. Using pharmacological and localized transgenic perturbations, 4D imaging of the zebrafish embryo, systematic analysis of cell motion, and computational modeling, we find that the vertebrate tail organizer orchestrates morphogenesis over distances beyond the range of morphogen signaling. The organizer regulates the rate and coherence of cell motion in the elongating embryo using mechanical information that is transmitted via relay between neighboring cells. This mechanism is similar to a pressure front in granular media and other jammed systems, but in the embryo the mechanical information emerges from self-propelled cell movement and not force transfer between cells. The propagation likely relies upon local biochemical signaling that affects cell contractility, cell adhesion, and/or cell polarity but is independent of transcription and translation.
Collapse
|
7
|
Mutational analysis of dishevelled genes in zebrafish reveals distinct functions in embryonic patterning and gastrulation cell movements. PLoS Genet 2018; 14:e1007551. [PMID: 30080849 PMCID: PMC6095615 DOI: 10.1371/journal.pgen.1007551] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/16/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
Wnt signaling plays critical roles in dorsoventral fate specification and anteroposterior patterning, as well as in morphogenetic cell movements. Dishevelled proteins, or Dvls, mediate the activation of Wnt/ß-catenin and Wnt/planar cell polarity pathways. There are at least three highly conserved Dvl proteins in vertebrates, but the implication of each Dvl in key early developmental processes remains poorly understood. In this study, we use genome-editing approach to generate different combinations of maternal and zygotic dvl mutants in zebrafish, and examine their functions during early development. Maternal transcripts for dvl2 and dvl3a are most abundantly expressed, whereas the transcript levels of other dvl genes are negligible. Phenotypic and molecular analyses show that early dorsal fate specification is not affected in maternal and zygotic dvl2 and dvl3a double mutants, suggesting that the two proteins may be dispensable for the activation of maternal Wnt/ß-catenin signaling. Interestingly, convergence and extension movements and anteroposterior patterning require both maternal and the zygotic functions of Dvl2 and Dvl3a, but these processes are more sensitive to Dvl2 dosage. Zygotic dvl2 and dvl3a double mutants display mild axis extension defect with correct anteroposterior patterning. However, maternal and zygotic double mutants exhibit most strongly impaired convergence and extension movements, severe trunk and posterior deficiencies, and frequent occurrence of cyclopia and craniofacial defects. Our results suggest that Dvl2 and Dvl3a products are required for the activation of zygotic Wnt/ß-catenin signaling and Wnt/planar cell polarity pathway, and regulate zygotic developmental processes in a dosage-dependent manner. This work provides insight into the mechanisms of Dvl-mediated Wnt signaling pathways during early vertebrate development. The embryogenesis of most animals is first supported by maternal gene products accumulated in the oocyte, and then by the expression of genes from the zygote. In all vertebrates, there are at least three Dishevelled (Dvl) proteins, which play critical roles in normal development and human diseases. They are both maternally and zygotically expressed, and can activate the ß-catenin-dependent Wnt pathway that regulates gene expression and cell fate, and the ß-catenin-independent Wnt pathway that orchestrates cell movements. In zebrafish embryo, Dvl2 and Dvl3a are most abundant, but their functions are not fully understood. We find that maternally and zygotically expressed Dvl2 plays a predominant role in the elongation of the anteroposterior axis, and the expression of genes involved in the development of the posterior region. Dvl3a cooperates with Dvl2 in these processes. Analyses after loss-of-function of these genes indicate that deficiency of maternal and zygotic Dvl2 and Dvl3a results in embryos with cyclopia, craniofacial defects, and severe abnormality in the trunk and posterior regions. Many human birth defects and other diseases, like cancer, are attributed to the dysfunction of the Wnt pathways. Our results help to understand the mechanisms of Dvl-mediated Wnt pathway activation, and the causes of developmental disorders.
Collapse
|
8
|
Kim HT, Lee MS, Jeong YM, Ro H, Kim DI, Shin YH, Kim JE, Hwang KS, Choi JH, Bahn M, Lee JJ, Lee SH, Bae YK, Lee JS, Choi JK, Kim NS, Yeo CY, Kim CH. Ottogi Inhibits Wnt/β-catenin Signaling by Regulating Cell Membrane Trafficking of Frizzled8. Sci Rep 2017; 7:13278. [PMID: 29038508 PMCID: PMC5643531 DOI: 10.1038/s41598-017-13429-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/22/2017] [Indexed: 01/30/2023] Open
Abstract
Wnt signaling controls critical developmental processes including tissue/body patterning. Here we report the identification of a novel regulator of Wnt signaling, OTTOGI (OTG), isolated from a large-scale expression screening of human cDNAs in zebrafish embryos. Overexpression of OTG in zebrafish embryos caused dorso-anteriorized phenotype, inhibited the expression of Wnt target genes, and prevented nuclear accumulation of β-catenin. Conversely, knockdown of zebrafish otg using specific antisense morpholino promoted nuclear accumulation of β-catenin and caused ventralization. However, OTG failed to rescue headless-like phenotype induced by inhibition of GSK-3β activity, suggesting that OTG acts upstream of GSK-3β. OTG bound specifically to Frizzled8 (Fz8) receptor and caused retention of Fz8 in the endoplasmic reticulum possibly by preventing N-linked glycosylation of Fz8. Taken together, our data indicate that OTG functions as a novel negative regulator of Wnt signaling during development by the modulation of cell surface expression of Fz receptor.
Collapse
Affiliation(s)
- Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Mi-Sun Lee
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Yun-Mi Jeong
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Hyunju Ro
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Dong-Il Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Yong-Hwan Shin
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Ji-Eun Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Kyu-Seok Hwang
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Minjin Bahn
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 120-750, South Korea
| | - Jeong-Ju Lee
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea
| | - Sang H Lee
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Young-Ki Bae
- National Cancer Center, Goyang, 410-769, South Korea
| | - Jin-Soo Lee
- National Cancer Center, Goyang, 410-769, South Korea
| | - Joong-Kook Choi
- Department of Biochemistry, College of Medicine, Chungbuk National University, Cheongju, 361-763, South Korea
| | - Nam-Soon Kim
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea.
| | - Chang-Yeol Yeo
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 120-750, South Korea.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
9
|
Morrow ZT, Maxwell AM, Hoshijima K, Talbot JC, Grunwald DJ, Amacher SL. tbx6l and tbx16 are redundantly required for posterior paraxial mesoderm formation during zebrafish embryogenesis. Dev Dyn 2017; 246:759-769. [PMID: 28691257 DOI: 10.1002/dvdy.24547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/19/2017] [Accepted: 07/04/2017] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND T-box genes encode a large transcription factor family implicated in many aspects of development. We are focusing on two related zebrafish T-box genes, tbx6l and tbx16, that are expressed in highly overlapping patterns in embryonic paraxial mesoderm. tbx16 mutants are deficient in trunk, but not tail, somites; we explored whether presence of tail somites in tbx16 mutants was due to compensatory function provided by the tbx6l gene. RESULTS We generated two zebrafish tbx6l mutant alleles. Loss of tbx6l has no apparent effect on embryonic development, nor does tbx6l loss enhance the phenotype of two other T-box gene mutants, ta and tbx6, or of the mesp family gene mutant msgn1. In contrast, loss of tbx6l function dramatically enhances the paraxial mesoderm deficiency of tbx16 mutants. CONCLUSIONS These data demonstrate that tbx6l and tbx16 genes function redundantly to direct tail somite development. tbx6l single mutants develop normally because tbx16 fully compensates for loss of tbx6l function. However, tbx6l only partially compensates for loss of tbx16 function. These results resolve the question of why loss of function of tbx16 gene, which is expressed throughout the ventral and paraxial mesoderm, profoundly affects somite development in the trunk but not the tail. Developmental Dynamics 246:759-769, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zachary T Morrow
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Adrienne M Maxwell
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Kazuyuki Hoshijima
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jared C Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio.,Department of Biological Chemistry and Pharmacology, The Ohio State University School of Medicine, Columbus, Ohio.,Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, Ohio
| | - David J Grunwald
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Sharon L Amacher
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio.,Department of Biological Chemistry and Pharmacology, The Ohio State University School of Medicine, Columbus, Ohio.,Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
10
|
Abstract
Cytokines of the transforming growth factor β (TGF-β) family, including TGF-βs, bone morphogenic proteins (BMPs), activins, and Nodal, play crucial roles in embryonic development and adult tissue homeostasis by regulating cell proliferation, survival, and differentiation, as well as stem-cell self-renewal and lineage-specific differentiation. Smad proteins are critical downstream mediators of these signaling activities. In addition to regulating the transcription of direct target genes of TGF-β, BMP, activin, or Nodal, Smad proteins also participate in extensive cross talk with other signaling pathways, often in a cell-type- or developmental stage-specific manner. These combinatorial signals often produce context-, time-, and location-dependent biological outcomes that are critical for development. This review discusses recent progress in our understanding of the cross talk between Smad proteins and signaling pathways of Wnt, Notch, Hippo, Hedgehog (Hh), mitogen-activated protein (MAP), kinase, phosphoinositide 3-kinase (PI3K)-Akt, nuclear factor κB (NF-κB), and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways.
Collapse
Affiliation(s)
- Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, and Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
11
|
Luo K. Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol 2017. [PMID: 27836834 DOI: 10.1101/cshperspect] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines of the transforming growth factor β (TGF-β) family, including TGF-βs, bone morphogenic proteins (BMPs), activins, and Nodal, play crucial roles in embryonic development and adult tissue homeostasis by regulating cell proliferation, survival, and differentiation, as well as stem-cell self-renewal and lineage-specific differentiation. Smad proteins are critical downstream mediators of these signaling activities. In addition to regulating the transcription of direct target genes of TGF-β, BMP, activin, or Nodal, Smad proteins also participate in extensive cross talk with other signaling pathways, often in a cell-type- or developmental stage-specific manner. These combinatorial signals often produce context-, time-, and location-dependent biological outcomes that are critical for development. This review discusses recent progress in our understanding of the cross talk between Smad proteins and signaling pathways of Wnt, Notch, Hippo, Hedgehog (Hh), mitogen-activated protein (MAP), kinase, phosphoinositide 3-kinase (PI3K)-Akt, nuclear factor κB (NF-κB), and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways.
Collapse
Affiliation(s)
- Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, and Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
12
|
The guanine nucleotide exchange factor Net1 facilitates the specification of dorsal cell fates in zebrafish embryos by promoting maternal β-catenin activation. Cell Res 2016; 27:202-225. [PMID: 27910850 DOI: 10.1038/cr.2016.141] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/18/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Wnt/β-catenin signaling is essential for the initiation of dorsal-ventral patterning during vertebrate embryogenesis. Maternal β-catenin accumulates in dorsal marginal nuclei during cleavage stages, but its critical target genes essential for dorsalization are silent until mid-blastula transition (MBT). Here, we find that zebrafish net1, a guanine nucleotide exchange factor, is specifically expressed in dorsal marginal blastomeres after MBT, and acts as a zygotic factor to promote the specification of dorsal cell fates. Loss- and gain-of-function experiments show that the GEF activity of Net1 is required for the activation of Wnt/β-catenin signaling in zebrafish embryos and mammalian cells. Net1 dissociates and activates PAK1 dimers, and PAK1 kinase activation causes phosphorylation of S675 of β-catenin after MBT, which ultimately leads to the transcription of downstream target genes. In summary, our results reveal that Net1-regulated β-catenin activation plays a crucial role in the dorsal axis formation during zebrafish development.
Collapse
|
13
|
BMP and retinoic acid regulate anterior-posterior patterning of the non-axial mesoderm across the dorsal-ventral axis. Nat Commun 2016; 7:12197. [PMID: 27406002 PMCID: PMC4947171 DOI: 10.1038/ncomms12197] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 06/09/2016] [Indexed: 11/23/2022] Open
Abstract
Despite the fundamental importance of patterning along the dorsal–ventral (DV) and anterior–posterior (AP) axes during embryogenesis, uncertainty exists in the orientation of these axes for the mesoderm. Here we examine the origin and formation of the zebrafish kidney, a ventrolateral mesoderm derivative, and show that AP patterning of the non-axial mesoderm occurs across the classic gastrula stage DV axis while DV patterning aligns along the animal–vegetal pole. We find that BMP signalling acts early to establish broad anterior and posterior territories in the non-axial mesoderm while retinoic acid (RA) functions later, but also across the classic DV axis. Our data support a model in which RA on the dorsal side of the embryo induces anterior kidney fates while posterior kidney progenitors are protected ventrally by the RA-catabolizing enzyme Cyp26a1. This work clarifies our understanding of vertebrate axis orientation and establishes a new paradigm for how the kidney and other mesodermal derivatives arise during embryogenesis. It is unclear how the dorsal-ventral (DV) and anterior-posterior (AP) axes established in the gastrula affect tissues. Here, the authors show that in zebrafish kidney development, with regard to non-axial mesoderm, the classic DV axis corresponds to the AP axis, and is regulated by BMP and retinoic acid.
Collapse
|
14
|
Zfp703 Is a Wnt/β-Catenin Feedback Suppressor Targeting the β-Catenin/Tcf1 Complex. Mol Cell Biol 2016; 36:1793-802. [PMID: 27090637 DOI: 10.1128/mcb.01010-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/05/2016] [Indexed: 01/29/2023] Open
Abstract
The Wnt/β-catenin signaling pathway controls embryonic development and adult stem cell maintenance through the regulation of transcription. Failure to downregulate Wnt signaling can result in embryonic malformations and cancer, highlighting the important role of negative regulators of the pathway. The Wnt pathway activates several negative feedback targets, including axin2 and Dkk1, that function at different levels of the signaling cascade; however, none have been identified that directly target active β-catenin/Tcf1 transcriptional complexes. We show that Zfp703 is a Wnt target gene that inhibits Wnt/β-catenin activity in Wnt reporter assays and in Wnt-dependent mesoderm differentiation in embryonic stem cells. Zfp703 binds directly to Tcf1 to inhibit β-catenin/Tcf1 complex formation and does so independently of the Groucho/Tle transcriptional corepressor. We propose that Zfp703 is a novel feedback suppressor of Wnt/β-catenin signaling that functions by inhibiting the association of β-catenin with Tcf1 on Wnt response elements in target gene enhancers.
Collapse
|
15
|
Yang K, Wang X, Zhang H, Wang Z, Nan G, Li Y, Zhang F, Mohammed MK, Haydon RC, Luu HH, Bi Y, He TC. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. J Transl Med 2016; 96:116-36. [PMID: 26618721 PMCID: PMC4731283 DOI: 10.1038/labinvest.2015.144] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/06/2015] [Indexed: 02/07/2023] Open
Abstract
The canonical WNT/β-catenin signaling pathway governs a myriad of biological processes underlying the development and maintenance of adult tissue homeostasis, including regulation of stem cell self-renewal, cell proliferation, differentiation, and apoptosis. WNTs are secreted lipid-modified glycoproteins that act as short-range ligands to activate receptor-mediated signaling pathways. The hallmark of the canonical pathway is the activation of β-catenin-mediated transcriptional activity. Canonical WNTs control the β-catenin dynamics as the cytoplasmic level of β-catenin is tightly regulated via phosphorylation by the 'destruction complex', consisting of glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), the scaffold protein AXIN, and the tumor suppressor adenomatous polyposis coli (APC). Aberrant regulation of this signaling cascade is associated with varieties of human diseases, especially cancers. Over the past decade, significant progress has been made in understanding the mechanisms of canonical WNT signaling. In this review, we focus on the current understanding of WNT signaling at the extracellular, cytoplasmic membrane, and intracellular/nuclear levels, including the emerging knowledge of cross-talk with other pathways. Recent progresses in developing novel WNT pathway-targeted therapies will also be reviewed. Thus, this review is intended to serve as a refresher of the current understanding about the physiologic and pathogenic roles of WNT/β-catenin signaling pathway, and to outline potential therapeutic opportunities by targeting the canonical WNT pathway.
Collapse
Affiliation(s)
- Ke Yang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xin Wang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Department of Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Zhongliang Wang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guoxin Nan
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yasha Li
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Fugui Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Maryam K. Mohammed
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| | - Tong-Chuan He
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| |
Collapse
|
16
|
Cross-Scale Integrin Regulation Organizes ECM and Tissue Topology. Dev Cell 2015; 34:33-44. [PMID: 26096733 DOI: 10.1016/j.devcel.2015.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 03/23/2015] [Accepted: 04/29/2015] [Indexed: 11/23/2022]
Abstract
The diverse morphologies of animal tissues are underlain by different configurations of adherent cells and extracellular matrix (ECM). Here, we elucidate a cross-scale mechanism for tissue assembly and ECM remodeling involving Cadherin 2, the ECM protein Fibronectin, and its receptor Integrin α5. Fluorescence cross-correlation spectroscopy within the zebrafish paraxial mesoderm mesenchyme reveals a physical association between Integrin α5 on adjacent cell membranes. This Integrin-Integrin complex correlates with conformationally inactive Integrin. Cadherin 2 stabilizes both the Integrin association and inactive Integrin conformation. Thus, Integrin repression within the adherent mesenchymal interior of the tissue biases Fibronectin fibrillogenesis to the tissue surface lacking cell-cell adhesions. Along nascent somite boundaries, Cadherin 2 levels decrease, becoming anti-correlated with levels of Integrin α5. Simultaneously, Integrin α5 clusters and adopts the active conformation and then commences ECM assembly. This cross-scale regulation of Integrin activation organizes a stereotypic pattern of ECM necessary for vertebrate body elongation and segmentation.
Collapse
|
17
|
Shimizu N, Ishitani S, Sato A, Shibuya H, Ishitani T. Hipk2 and PP1c Cooperate to Maintain Dvl Protein Levels Required for Wnt Signal Transduction. Cell Rep 2014; 8:1391-404. [DOI: 10.1016/j.celrep.2014.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 06/04/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022] Open
|
18
|
Kirmizitas A, Gillis WQ, Zhu H, Thomsen GH. Gtpbp2 is required for BMP signaling and mesoderm patterning in Xenopus embryos. Dev Biol 2014; 392:358-67. [PMID: 24858484 DOI: 10.1016/j.ydbio.2014.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/27/2014] [Accepted: 05/07/2014] [Indexed: 12/26/2022]
Abstract
Smad proteins convey canonical intracellular signals for activated receptors in the TGFβ superfamily, but the activity of Smads and their impact on target genes are further regulated by a wide variety of cofactors and partner proteins. We have identified a new Smad1 partner, a GTPase named Gtpbp2 that is a distant relative of the translation factor eEf1a. Gtpbp2 affects canonical signaling in the BMP branch of the TGFβ superfamily, as morpholino knockdown of Gtpbp2 decreases, and overexpression of Gtpbp2 enhances, animal cap responses to BMP4. During Xenopus development, gtpbp2 transcripts are maternally expressed and localized to the egg animal pole, and partitioned into the nascent ectodermal and mesodermal cells during cleavage and early gastrulation stages. Subsequently, gtpbp2 is expressed in the neural folds, and in early tadpoles undergoing organogenesis gtpbp2 is expressed prominently in the brain, eyes, somites, ventral blood island and branchial arches. Consistent with its expression, morpholino knockdown of Gtpbp2 causes defects in ventral-posterior germ layer patterning, gastrulation and tadpole morphology. Overexpressed Gtpbp2 can induce ventral-posterior marker genes and localize to cell nuclei in Xenopus animal caps, highlighting its role in regulating BMP signaling in the early embryo. Here, we introduce this large GTPase as a novel factor in BMP signaling and ventral-posterior patterning.
Collapse
Affiliation(s)
- Arif Kirmizitas
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - William Q Gillis
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Haitao Zhu
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Gerald H Thomsen
- Department of Biochemistry and Cell Biology, Graduate Program in Molecular and Cellular Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
19
|
Bajard L, Morelli LG, Ares S, Pécréaux J, Jülicher F, Oates AC. Wnt-regulated dynamics of positional information in zebrafish somitogenesis. Development 2014; 141:1381-91. [PMID: 24595291 PMCID: PMC3943186 DOI: 10.1242/dev.093435] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
How signaling gradients supply positional information in a field of moving cells is an unsolved question in patterning and morphogenesis. Here, we ask how a Wnt signaling gradient regulates the dynamics of a wavefront of cellular change in a flow of cells during somitogenesis. Using time-controlled perturbations of Wnt signaling in the zebrafish embryo, we changed segment length without altering the rate of somite formation or embryonic elongation. This result implies specific Wnt regulation of the wavefront velocity. The observed Wnt signaling gradient dynamics and timing of downstream events support a model for wavefront regulation in which cell flow plays a dominant role in transporting positional information.
Collapse
Affiliation(s)
- Lola Bajard
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Zhao T, Gan Q, Stokes A, Lassiter RNT, Wang Y, Chan J, Han JX, Pleasure DE, Epstein JA, Zhou CJ. β-catenin regulates Pax3 and Cdx2 for caudal neural tube closure and elongation. Development 2013; 141:148-57. [PMID: 24284205 DOI: 10.1242/dev.101550] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-canonical Wnt/planar cell polarity (PCP) signaling plays a primary role in the convergent extension that drives neural tube closure and body axis elongation. PCP signaling gene mutations cause severe neural tube defects (NTDs). However, the role of canonical Wnt/β-catenin signaling in neural tube closure and NTDs remains poorly understood. This study shows that conditional gene targeting of β-catenin in the dorsal neural folds of mouse embryos represses the expression of the homeobox-containing genes Pax3 and Cdx2 at the dorsal posterior neuropore (PNP), and subsequently diminishes the expression of the Wnt/β-catenin signaling target genes T, Tbx6 and Fgf8 at the tail bud, leading to spina bifida aperta, caudal axis bending and tail truncation. We demonstrate that Pax3 and Cdx2 are novel downstream targets of Wnt/β-catenin signaling. Transgenic activation of Pax3 cDNA can rescue the closure defect in the β-catenin mutants, suggesting that Pax3 is a key downstream effector of β-catenin signaling in the PNP closure process. Cdx2 is known to be crucial in posterior axis elongation and in neural tube closure. We found that Cdx2 expression is also repressed in the dorsal PNPs of Pax3-null embryos. However, the ectopically activated Pax3 in the β-catenin mutants cannot restore Cdx2 mRNA in the dorsal PNP, suggesting that the presence of both β-catenin and Pax3 is required for regional Cdx2 expression. Thus, β-catenin signaling is required for caudal neural tube closure and elongation, acting through the transcriptional regulation of key target genes in the PNP.
Collapse
Affiliation(s)
- Tianyu Zhao
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Steinfeld J, Steinfeld I, Coronato N, Hampel ML, Layer PG, Araki M, Vogel-Höpker A. RPE specification in the chick is mediated by surface ectoderm-derived BMP and Wnt signalling. Development 2013; 140:4959-69. [PMID: 24227655 DOI: 10.1242/dev.096990] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The retinal pigment epithelium (RPE) is indispensable for vertebrate eye development and vision. In the classical model of optic vesicle patterning, the surface ectoderm produces fibroblast growth factors (FGFs) that specify the neural retina (NR) distally, whereas TGFβ family members released from the proximal mesenchyme are involved in RPE specification. However, we previously proposed that bone morphogenetic proteins (BMPs) released from the surface ectoderm are essential for RPE specification in chick. We now show that the BMP- and Wnt-expressing surface ectoderm is required for RPE specification. We reveal that Wnt signalling from the overlying surface ectoderm is involved in restricting BMP-mediated RPE specification to the dorsal optic vesicle. Wnt2b is expressed in the dorsal surface ectoderm and subsequently in dorsal optic vesicle cells. Activation of Wnt signalling by implanting Wnt3a-soaked beads or inhibiting GSK3β at optic vesicle stages inhibits NR development and converts the entire optic vesicle into RPE. Surface ectoderm removal at early optic vesicle stages or inhibition of Wnt, but not Wnt/β-catenin, signalling prevents pigmentation and downregulates the RPE regulatory gene Mitf. Activation of BMP or Wnt signalling can replace the surface ectoderm to rescue MITF expression and optic cup formation. We provide evidence that BMPs and Wnts cooperate via a GSK3β-dependent but β-catenin-independent pathway at the level of pSmad to ensure RPE specification in dorsal optic vesicle cells. We propose a new dorsoventral model of optic vesicle patterning, whereby initially surface ectoderm-derived Wnt signalling directs dorsal optic vesicle cells to develop into RPE through a stabilising effect of BMP signalling.
Collapse
Affiliation(s)
- Jörg Steinfeld
- Fachgebiet Entwicklungsbiologie und Neurogenetik, Technische Universität Darmstadt, Schnittspahnstrasse 13, D-64287 Darmstadt, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Wang S, Yin J, Chen D, Nie F, Song X, Fei C, Miao H, Jing C, Ma W, Wang L, Xie S, Li C, Zeng R, Pan W, Hao X, Li L. Small-molecule modulation of Wnt signaling via modulating the Axin-LRP5/6 interaction. Nat Chem Biol 2013; 9:579-85. [PMID: 23892894 DOI: 10.1038/nchembio.1309] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 06/26/2013] [Indexed: 01/07/2023]
Abstract
The Wnt/β-catenin signaling pathway has a crucial role in embryonic development, stem cell maintenance and human disease. By screening a synthetic chemical library of lycorine derivatives, we identified 4-ethyl-5-methyl-5,6-dihydro-[1,3]dioxolo[4,5-j]phenanthridine (HLY78) as an activator of the Wnt/β-catenin signaling pathway, which acts in a Wnt ligand-dependent manner. HLY78 targets the DIX domain of Axin and potentiates the Axin-LRP6 association, thus promoting LRP6 phosphorylation and Wnt signaling transduction. Moreover, we identified the critical residues on Axin for HLY78 binding and showed that HLY78 may weaken the autoinhibition of Axin. In addition, HLY78 acts synergistically with Wnt in the embryonic development of zebrafish and increases the expression of the conserved hematopoietic stem cell (HSC) markers, runx1 and cmyb, in zebrafish embryos. Collectively, our study not only provides new insights into the regulation of the Wnt/β-catenin signaling pathway by a Wnt-specific small molecule but also will facilitate therapeutic applications, such as HSC expansion.
Collapse
Affiliation(s)
- Sheng Wang
- 1] State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China. [2]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dray N, Lawton A, Nandi A, Jülich D, Emonet T, Holley SA. Cell-fibronectin interactions propel vertebrate trunk elongation via tissue mechanics. Curr Biol 2013; 23:1335-41. [PMID: 23810535 DOI: 10.1016/j.cub.2013.05.052] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/07/2013] [Accepted: 05/28/2013] [Indexed: 11/27/2022]
Abstract
During embryonic development and tissue homeostasis, cells produce and remodel the extracellular matrix (ECM). The ECM maintains tissue integrity and can serve as a substrate for cell migration. Integrin α5 (Itgα5) and αV (ItgαV) are the α subunits of the integrins most responsible for both cell adhesion to the ECM protein fibronectin (FN) and FN matrix fibrillogenesis. We perform a systems-level analysis of cell motion in the zebrafish tail bud during trunk elongation in the presence and absence of normal cell-FN interactions. Itgα5 and ItgαV have well-described roles in cell migration in vitro. However, we find that concomitant loss of itgα5 and itgαV leads to a trunk elongation defect without substantive alteration of cell migration. Tissue-specific transgenic rescue experiments suggest that the FN matrix on the surface of the paraxial mesoderm is required for body elongation via its role in defining tissue mechanics and intertissue adhesion.
Collapse
Affiliation(s)
- Nicolas Dray
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
25
|
Lee RTH, Knapik EW, Thiery JP, Carney TJ. An exclusively mesodermal origin of fin mesenchyme demonstrates that zebrafish trunk neural crest does not generate ectomesenchyme. Development 2013; 140:2923-32. [PMID: 23739134 DOI: 10.1242/dev.093534] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The neural crest is a multipotent stem cell population that arises from the dorsal aspect of the neural tube and generates both non-ectomesenchymal (melanocytes, peripheral neurons and glia) and ectomesenchymal (skeletogenic, odontogenic, cartilaginous and connective tissue) derivatives. In amniotes, only cranial neural crest generates both classes, with trunk neural crest restricted to non-ectomesenchyme. By contrast, it has been suggested that anamniotes might generate derivatives of both classes at all axial levels, with trunk neural crest generating fin osteoblasts, scale mineral-forming cells and connective tissue cells; however, this has not been fully tested. The cause and evolutionary significance of this cranial/trunk dichotomy, and its absence in anamniotes, are debated. Recent experiments have disputed the contribution of fish trunk neural crest to fin osteoblasts and scale mineral-forming cells. This prompted us to test the contribution of anamniote trunk neural crest to fin connective tissue cells. Using genetics-based lineage tracing in zebrafish, we find that these fin mesenchyme cells derive entirely from the mesoderm and that neural crest makes no contribution. Furthermore, contrary to previous suggestions, larval fin mesenchyme cells do not generate the skeletogenic cells of the adult fin, but persist to form fibroblasts associated with adult fin rays. Our data demonstrate that zebrafish trunk neural crest does not generate ectomesenchymal derivatives and challenge long-held ideas about trunk neural crest fate. These findings have important implications for the ontogeny and evolution of the neural crest.
Collapse
Affiliation(s)
- Raymond Teck Ho Lee
- Institute of Molecular and Cell Biology-IMCB, A*STAR-Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | | | | | | |
Collapse
|
26
|
O'Neill K, Thorpe C. BMP signaling and spadetail regulate exit of muscle precursors from the zebrafish tailbud. Dev Biol 2013; 375:117-27. [PMID: 23246591 DOI: 10.1016/j.ydbio.2012.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 11/26/2012] [Accepted: 12/04/2012] [Indexed: 11/19/2022]
Abstract
The tailbud is a population of stem cells in the posterior embryonic tail. During zebrafish development, these stem cells give rise to the main structures of the embryo's posterior body, including the tail somites. Progenitor cells reside in the tailbud for variable amounts of time before they exit and begin to differentiate. There must be a careful balance between cells that leave the tailbud and cells that are held back in order to give rise to later somites. However, this meticulous process is not well understood. A gene that has shed some light on this area is the t-box transcription factor spadetail (spt). When spt is mutated, embryos develop an enlarged tailbud and are only able to form roughly half of their somites. This phenotype is due to the fact that some of the somitic precursors are not able to leave the tailbud or differentiate. Another factor involved in tail morphogenesis is the Bone Morphogenetic Protein (BMP) pathway. BMPs are important for many processes during early development, including cell migration. Chordino (chd) is a secreted protein that inhibits BMP signaling. BMPs are upregulated in chd mutants, however, these mutants are able to form organized somites. In embryos where chd and spt are mutated, somites are completely absent. These double mutants also develop a large tailbud due to the accumulation of progenitor cells that are never able to leave or differentiate. To study the dynamics of cells in the tailbud and their role in somite formation, we have analyzed the genetic factors and pathway interactions involved, conducted transplant experiments to look at behavior of mutant cells in different genetic backgrounds, and used time lapse microscopy to characterize cell movements and behavior in wild type and mutant tailbuds. These data suggest that spt expression and BMP inhibition are both required for somitic precursors to exit the tailbud. They also elucidate that chd;spt tailbud mesodermal progenitor cells (MPC) behave autonomously and their dynamics within the tailbud are drastically different than WT MPCs.
Collapse
Affiliation(s)
- Katelyn O'Neill
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | | |
Collapse
|
27
|
Lawton AK, Nandi A, Stulberg MJ, Dray N, Sneddon MW, Pontius W, Emonet T, Holley SA. Regulated tissue fluidity steers zebrafish body elongation. Development 2013; 140:573-82. [PMID: 23293289 DOI: 10.1242/dev.090381] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The tailbud is the posterior leading edge of the growing vertebrate embryo and consists of motile progenitors of the axial skeleton, musculature and spinal cord. We measure the 3D cell flow field of the zebrafish tailbud and identify changes in tissue fluidity revealed by reductions in the coherence of cell motion without alteration of cell velocities. We find a directed posterior flow wherein the polarization between individual cell motion is high, reflecting ordered collective migration. At the posterior tip of the tailbud, this flow makes sharp bilateral turns facilitated by extensive cell mixing due to increased directional variability of individual cell motions. Inhibition of Wnt or Fgf signaling or cadherin 2 function reduces the coherence of the flow but has different consequences for trunk and tail extension. Modeling and additional data analyses suggest that the balance between the coherence and rate of cell flow determines whether body elongation is linear or whether congestion forms within the flow and the body axis becomes contorted.
Collapse
Affiliation(s)
- Andrew K Lawton
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Liu JX, Zhang D, Xie X, Ouyang G, Liu X, Sun Y, Xiao W. Eaf1 and Eaf2 negatively regulate canonical Wnt/β-catenin signaling. Development 2013; 140:1067-78. [PMID: 23364330 DOI: 10.1242/dev.086157] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Eaf factors play a crucial role in tumor suppression and embryogenesis. To investigate the potential mechanism of Eaf activity, we performed loss- and gain-of-function assays in zebrafish using morpholino and mRNA injections, respectively. We found that eaf1 and eaf2 inhibit Wnt/β-catenin signaling, thereby modulating mesodermal and neural patterning in the embryo. Moreover, ectopic expression of eaf1 and eaf2 in embryos and cultured cells blocked β-catenin reporter activity. By immunoprecipitation, we also observed that Eaf1 and Eaf2 bound to the Armadillo repeat region and C-terminus of β-catenin, as well as to other β-catenin transcription complex proteins, such as c-Jun, Tcf and Axin, suggesting the formation of a novel complex. In addition, the N-terminus of Eaf1 and Eaf2 bound to β-catenin and exhibited dominant-negative activity, whereas the C-terminus appeared to either harbor a suppression domain or to recruit a repressor. Both the N- and C-terminus must be intact for Eaf1 and Eaf2 suppressive activity. Lastly, we demonstrate a conservation of biological activities for Eaf family proteins across species. In summary, our evidence points to a novel role for Eaf1 and Eaf2 in inhibiting canonical Wnt/β-catenin signaling, which might form the mechanistic basis for Eaf1 and Eaf2 tumor suppressor activity.
Collapse
Affiliation(s)
- Jing-Xia Liu
- Key Laboratory of Biodiversity and Conservation of Aquatic Organisms, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, PR China
| | | | | | | | | | | | | |
Collapse
|
29
|
Stulberg MJ, Lin A, Zhao H, Holley SA. Crosstalk between Fgf and Wnt signaling in the zebrafish tailbud. Dev Biol 2012; 369:298-307. [PMID: 22796649 PMCID: PMC3423502 DOI: 10.1016/j.ydbio.2012.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 06/26/2012] [Accepted: 07/02/2012] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor (Fgf) and Wnt signaling are necessary for the intertwined processes of tail elongation, mesodermal development and somitogenesis. Here, we use pharmacological modifiers and time-resolved quantitative analysis of both nascent transcription and protein phosphorylation in the tailbud, to distinguish early effects of signal perturbation from later consequences related to cell fate changes. We demonstrate that Fgf activity elevates Wnt signaling by inhibiting transcription of the Wnt antagonists dkk1 and notum1a. PI3 kinase signaling also increases Wnt signaling via phosphorylation of Gsk3β. Conversely, Wnt can increase signaling within the Mapk branch of the Fgf pathway as Gsk3β phosphorylation elevates phosphorylation levels of Erk. Despite the reciprocal positive regulation between Fgf and Wnt, the two pathways generally have opposing effects on the transcription of co-regulated genes. This opposing regulation of target genes may represent a rudimentary relationship that manifests as out-of-phase oscillation of Fgf and Wnt target genes in the mouse and chick tailbud. In summary, these data suggest that Fgf and Wnt signaling are tightly integrated to maintain proportional levels of activity in the zebrafish tailbud, and this balance is important for axis elongation, cell fate specification and somitogenesis.
Collapse
Affiliation(s)
- Michael J. Stulberg
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Aiping Lin
- Keck Biostatistics Resource, Yale University, New Haven, CT 06511, USA
| | - Hongyu Zhao
- Keck Biostatistics Resource, Yale University, New Haven, CT 06511, USA
- Department of Epidemiology and Public Health, Yale University, New Haven, CT 06511, USA
| | - Scott A. Holley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
30
|
Shimizu N, Kawakami K, Ishitani T. Visualization and exploration of Tcf/Lef function using a highly responsive Wnt/β-catenin signaling-reporter transgenic zebrafish. Dev Biol 2012; 370:71-85. [PMID: 22842099 DOI: 10.1016/j.ydbio.2012.07.016] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 07/03/2012] [Accepted: 07/10/2012] [Indexed: 01/09/2023]
Abstract
Evolutionarily conserved Tcf/Lef transcription factors (Lef1, Tcf7, Tcf7l1, and Tcf7l2) mediate gene expression regulated by Wnt/β-catenin signaling, which has multiple roles in early embryogenesis, organogenesis, adult tissue homeostasis, and tissue regeneration. However, the spatiotemporal dynamics of Tcf/Lef activity during these events remain poorly understood. We generated stable transgenic zebrafish lines carrying a new Wnt/β-catenin signaling reporter, Tcf/Lef-miniP:dGFP. The reporter revealed the transcriptional activities of four Tcf/Lef members controlled by Wnt/β-catenin signaling, which were expressed in known Wnt/β-catenin signaling-active sites during embryogenesis, organ development and growth, and tissue regeneration. We used the transgenic lines to demonstrate the contribution of Tcf/Lef-mediated Wnt/β-catenin signaling to the development of the anterior lateral line, dorsal and secondary posterior lateral lines, and gill filaments. Thus, these reporter lines are highly useful tools for studying Tcf/Lef-mediated Wnt/β-catenin signaling-dependent processes.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Division of Cell Regulation Systems, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | |
Collapse
|
31
|
Deng YZ, Yao F, Li JJ, Mao ZF, Hu PT, Long LY, Li G, Ji XD, Shi S, Guan DX, Feng YY, Cui L, Li DS, Liu Y, Du X, Guo MZ, Xu LY, Li EM, Wang HY, Xie D. RACK1 suppresses gastric tumorigenesis by stabilizing the β-catenin destruction complex. Gastroenterology 2012; 142:812-823.e15. [PMID: 22240482 DOI: 10.1053/j.gastro.2011.12.046] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 12/12/2011] [Accepted: 12/31/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Dysregulation of Wnt signaling has been involved in gastric tumorigenesis by mechanisms that are not fully understood. The receptor for activated protein kinase C (RACK1, GNB2L1) is involved in development of different tumor types, but its expression and function have not been investigated in gastric tumors. METHODS We analyzed expression of RACK1 in gastric tumor samples and their matched normal tissues from 116 patients using immunohistochemistry. Effects of knockdown with small interfering RNAs or overexpression of RACK1 in gastric cancer cell lines were evaluated in cell growth and tumor xenograft. RACK1 signaling pathways were investigated in cells and zebrafish embryos using immunoblot, immunoprecipitation, microinjection, and in situ hybridization assays. RESULTS Expression of RACK1 was reduced in gastric tumor samples and correlated with depth of tumor infiltration and poor differentiation. Knockdown of RACK1 in gastric cancer cells accelerated their anchorage-independent proliferation in soft agar, whereas overexpression of RACK1 reduced their tumorigenicity in nude mice. RACK1 formed a complex with glycogen synthase kinase Gsk3β and Axin to promote the interaction between Gsk3β and β-catenin and thereby stabilized the β-catenin destruction complex. On stimulation of Wnt3a, RACK1 repressed Wnt signaling by inhibiting recruitment of Axin by Dishevelled 2 (Dvl2). Moreover, there was an inverse correlation between expression of RACK1 and localization of β-catenin to the cytoplasm/nucleus in human gastric tumor samples. CONCLUSIONS RACK1 negatively regulates Wnt signaling pathway by stabilizing the β-catenin destruction complex and act as a tumor suppressor in gastric cancer cells.
Collapse
Affiliation(s)
- Yue-Zhen Deng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Martin BL, Kimelman D. Canonical Wnt signaling dynamically controls multiple stem cell fate decisions during vertebrate body formation. Dev Cell 2012; 22:223-32. [PMID: 22264734 PMCID: PMC3465166 DOI: 10.1016/j.devcel.2011.11.001] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/23/2011] [Accepted: 11/01/2011] [Indexed: 12/11/2022]
Abstract
The vertebrate body forms in an anterior-to-posterior progression, driven by a population of undifferentiated cells at the posterior-most end of the embryo. Recent studies have demonstrated that these undifferentiated cells are multipotent stem cells, suggesting that local signaling factors specify cell fate. However, the mechanism of cell fate specification during this process is unknown. Using a combination of single cell transplantation and newly developed cell-autonomous inducible Wnt inhibitor and activator transgenic zebrafish lines, we show that canonical Wnt signaling is continuously necessary and sufficient to specify mesoderm from a bipotential neural/mesodermal precursor. Surprisingly, we also find that Wnt signaling functions subsequently within the mesoderm to specify somites instead of posterior vascular endothelium. Our results demonstrate that dynamic local Wnt signaling cues specify germ layer contribution and mesodermal tissue type specification of multipotent stem cells throughout the formation of the early vertebrate embryonic body.
Collapse
Affiliation(s)
- Benjamin L. Martin
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Kimelman
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
33
|
Xie XW, Liu JX, Hu B, Xiao W. Zebrafish foxo3b negatively regulates canonical Wnt signaling to affect early embryogenesis. PLoS One 2011; 6:e24469. [PMID: 21915332 PMCID: PMC3168510 DOI: 10.1371/journal.pone.0024469] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/10/2011] [Indexed: 01/31/2023] Open
Abstract
FOXO genes are involved in many aspects of development and vascular homeostasis by regulating cell apoptosis, proliferation, and the control of oxidative stress. In addition, FOXO genes have been showed to inhibit Wnt/β-catenin signaling by competing with T cell factor to bind to β-catenin. However, how important of this inhibition in vivo, particularly in embryogenesis is still unknown. To demonstrate the roles of FOXO genes in embryogenesis will help us to further understand their relevant physiological functions. Zebrafish foxo3b gene, an orthologue of mammalian FOXO3, was expressed maternally and distributed ubiquitously during early embryogenesis and later restricted to brain. After morpholino-mediated knockdown of foxo3b, the zebrafish embryos exhibited defects in axis and neuroectoderm formation, suggesting its critical role in early embryogenesis. The embryo-developmental marker gene staining at different stages, phenotype analysis and rescue assays revealed that foxo3b acted its role through negatively regulating both maternal and zygotic Wnt/β-catenin signaling. Moreover, we found that foxo3b could interact with zebrafish β-catenin1 and β-catenin2 to suppress their transactivation in vitro and in vivo, further confirming its role relevant to the inhibition of Wnt/β-catenin signaling. Taken together, we revealed that foxo3b played a very important role in embryogenesis and negatively regulated maternal and zygotic Wnt/β-catenin signaling by directly interacting with both β-catenin1 and β-catenin2. Our studies provide an in vivo model for illustrating function of FOXO transcription factors in embryogenesis.
Collapse
Affiliation(s)
- Xun-wei Xie
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Jing-Xia Liu
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Bo Hu
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Wuhan Xiao
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Howard S, Deroo T, Fujita Y, Itasaki N. A positive role of cadherin in Wnt/β-catenin signalling during epithelial-mesenchymal transition. PLoS One 2011; 6:e23899. [PMID: 21909376 PMCID: PMC3166074 DOI: 10.1371/journal.pone.0023899] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 07/27/2011] [Indexed: 01/18/2023] Open
Abstract
The Wnt/β-catenin signalling pathway shares a key component, β-catenin, with the cadherin-based adhesion system. The signalling function of β-catenin is conferred by a soluble cytoplasmic pool that is unstable in the absence of a Wnt signal, whilst the adhesion function is based on a cadherin-bound, stable pool at the membrane. The cadherin complex is dynamic, allowing for cell-cell rearrangements such as epithelial-mesenchymal transition (EMT), where the complex turns over through internalisation. Potential interplay between the two pools remains poorly understood, but cadherins are generally considered negative regulators of Wnt signalling because they sequester cytoplasmic β-catenin. Here we explore how cellular changes at EMT affect the signalling capacity of β-catenin using two models of EMT: hepatocyte growth factor (HGF) treatment of MDCK cells, and gastrulation in embryonic development. We show that EMT not only provides a pool of signalling-competent β-catenin following internalisation of cadherin, but also significantly facilitates activation of the Wnt pathway in response to both Wnt signals and exogenous β-catenin. We further demonstrate that availability of β-catenin in the cytoplasm does not necessarily correlate with Wnt/β-catenin pathway activity, since blocking endocytosis or depleting endogenous cadherin abolishes pathway activation despite the presence of β-catenin in the cytoplasm. Lastly we present data suggesting that cadherins are required for augmented activation of the Wnt/β-catenin pathway in vivo. This suggests that cadherins play a crucial role in β-catenin-dependent transcription.
Collapse
Affiliation(s)
- Sara Howard
- MRC National Institute for Medical Research, London, United Kingdom
| | - Tom Deroo
- MRC National Institute for Medical Research, London, United Kingdom
| | - Yasuyuki Fujita
- MRC Laboratory for Molecular Cell Biology and Cell Biology Unit, Department of Cell and Developmental Biology, University College London, London, United Kingdom
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Nobue Itasaki
- MRC National Institute for Medical Research, London, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Hu S, Wu Z, Yan Y, Li Y. Sox31 is involved in central nervous system anteroposterior regionalization through regulating the organizer activity in zebrafish. Acta Biochim Biophys Sin (Shanghai) 2011; 43:387-99. [PMID: 21467072 DOI: 10.1093/abbs/gmr025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Sox superfamily proteins are DNA-binding transcriptional factors that contain highly conserved high-mobility group (HMG) box and take part in various development process. Sox31 is a maternal factor supplied in the oocyte and starts its zygotic expression during mid-blastula transition (MBT). From gastrulation stage, it mainly resides in neural tissue. Ectopically expression of Sox31 mRNA leads to cyclopia, fusion eyes, or totally loss of anterior head structure, in accompany with severe notochord defects. Molecular markers indicate that forebrain tissue reduces sharply while the posterior neural tissue expands anteriorly. In addition, organizer specification is also suppressed. Oppositely, an antisense morpholino designed functionally knockdown Sox31 causes typically dorsalized phenotype and reversed central nervous system (CNS) anteroposterior (AP) patterning. Gain of function with chimeric construct, where Sox31 HMG DNA binding domain is fused to a transcription activation domain (VP16) or transcription suppression domain (EnR), suggests that Sox31 acts as a transcriptional suppressor in vivo. The expression of Bozozok (Dharma), a direct target gene of pre-MBT Wnt/β-catenin signal, is suppressed by Sox31. Thus, to unveil the relationship between Sox31 and β-catenin-related transcriptional activity, we designed Top/Fop luciferase assay in HEK293T cells, and found that Sox31 could indeed suppress Tcf/Lef-dependent transcriptional activity without influencing the stability of β-catenin. Moreover, post-MBT Wnt signal was reduced in Sox31 morphants corresponding to the suppressed hindbrain structure, while phenotypic defects caused by excessive Sox31 could be rescued by Wnt antagonist dkk1. Taken together, Sox31 functions as an essential CNS AP patterning determinant and coordinates the CNS AP patterning process with organizer specification.
Collapse
Affiliation(s)
- Shengnan Hu
- Laboratory of Molecular Cell Biology, Shanghai Key Laboratory for Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | |
Collapse
|
36
|
Seebald JL, Szeto DP. Zebrafish eve1 regulates the lateral and ventral fates of mesodermal progenitor cells at the onset of gastrulation. Dev Biol 2011; 349:78-89. [DOI: 10.1016/j.ydbio.2010.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 10/01/2010] [Accepted: 10/05/2010] [Indexed: 12/13/2022]
|
37
|
Nakano N, Itoh S, Watanabe Y, Maeyama K, Itoh F, Kato M. Requirement of TCF7L2 for TGF-beta-dependent transcriptional activation of the TMEPAI gene. J Biol Chem 2010; 285:38023-33. [PMID: 20889500 DOI: 10.1074/jbc.m110.132209] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The TGF-β and Wnt pathways are involved in cell fate and tumorigenicity. A recent report indicated that a TGF-β target gene, TMEPAI (transmembrane prostate androgen-induced RNA), is possibly also a downstream target of Wnt signaling. Although TMEPAI was believed to be involved in tumorigenicity because of its blockage of TGF-β signaling, how TGF-β and Wnt signals affect the activation of the TMEPAI gene is not well understood. Herein, we show that the TMEPAI promoter is regulated synergistically by TGF-β/Smad and Wnt/β-catenin/T cell factor (TCF) 7L2. The critical cis-element for dual signals, termed TGF-β-responsive TCF7L2-binding element (TTE), is located in intron 1 of the TMEPAI gene. TCF7L2, but not Smad proteins, bound to TTE, whereas the disruption of TTE by mutagenesis remarkably counteracted both TGF-β and TCF7L2 responses. The introduction of mutations in critical Smad-binding elements blocked the activation of the TMEPAI promoter by TCF7L2. Furthermore, our DNA-protein interaction experiments revealed the indirect binding of TCF7L2 to Smad-binding elements via Smad3 upon TGF-β stimulation as well as its TGF-β-dependent association with TTE. We demonstrate that the Wnt/β-catenin/TCF7L2 pathway is preferentially able to alter the transcriptional regulation of the TGF-β-target gene, TMEPAI.
Collapse
Affiliation(s)
- Naoko Nakano
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Baker KD, Ramel MC, Lekven AC. A direct role for Wnt8 in ventrolateral mesoderm patterning. Dev Dyn 2010; 239:2828-36. [DOI: 10.1002/dvdy.22419] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
39
|
Mueller RL, Huang C, Ho RK. Spatio-temporal regulation of Wnt and retinoic acid signaling by tbx16/spadetail during zebrafish mesoderm differentiation. BMC Genomics 2010; 11:492. [PMID: 20828405 PMCID: PMC2996988 DOI: 10.1186/1471-2164-11-492] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 09/09/2010] [Indexed: 01/02/2023] Open
Abstract
Background A complex network of signaling pathways and transcription factors regulates vertebrate mesoderm development. Zebrafish mutants provide a powerful tool for examining the roles of individual genes in such a network. spadetail (spt) is a mutant with a lesion in tbx16, a T-box transcription factor involved in mesoderm development; the mutant phenotype includes disrupted primitive red blood cell formation as well as disrupted somitogenesis. Despite much recent progress, the downstream targets of tbx16 remain incompletely understood. The current study was carried out to test whether any of the five major signaling pathways are regulated by tbx16 during two specific stages of mesoderm development: primitive red blood cell formation in the intermediate mesoderm and somite formation in the tail paraxial mesoderm. This test was performed using Gene Set Enrichment Analysis, which identifies coordinated changes in expression among a priori sets of genes associated with biological features or processes. Results Our Gene Set Enrichment Analysis results identify Wnt and retinoic acid signaling as likely downstream targets of tbx16 in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation. In addition, such results identify retinoic acid signaling as a downstream target of tbx16 in the developing zebrafish posterior somites. Finally, using candidate gene identification and in situ hybridization, we provide expression domain information for 25 additional genes downstream of tbx16 that are outside of both pathways; 23 were previously unknown downstream targets of tbx16, and seven had previously uncharacterized expression in zebrafish. Conclusions Our results suggest that (1) tbx16 regulates Wnt signaling in the developing zebrafish intermediate mesoderm, the site of primitive red blood cell formation, and (2) tbx16 regulates retinoic acid signaling at two distinct embryonic locations and developmental stages, which may imply ongoing spatio-temporal regulation throughout mesoderm development.
Collapse
Affiliation(s)
- Rachel Lockridge Mueller
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
40
|
Caveolin-1 regulates dorsoventral patterning through direct interaction with β-catenin in zebrafish. Dev Biol 2010; 344:210-23. [DOI: 10.1016/j.ydbio.2010.04.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 04/28/2010] [Accepted: 04/29/2010] [Indexed: 01/22/2023]
|
41
|
Wang Y, Seebald JL, Szeto DP, Irudayaraj J. Biocompatibility and biodistribution of surface-enhanced Raman scattering nanoprobes in zebrafish embryos: in vivo and multiplex imaging. ACS NANO 2010; 4:4039-53. [PMID: 20552995 DOI: 10.1021/nn100351h] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nanoparticles are increasingly being used to investigate biological processes in various animal models due to their versatile chemical, unique optical, and multifunctional properties. In this report we address the biocompatibility and biodistribution of nanoparticle sensors used for Raman chemical imaging in live zebrafish (Danio rerio) embryos. Surface-enhanced Raman scattering (SERS) nanoprobes (NPs) comprising gold nanoparticles (AuNPs) as enhancing substrate and nonfluorescent Raman labels were synthesized and microinjected into zebrafish embryos at the one-cell stage. Raman mapping was performed to assess their distribution in various cell-types and tissues of developing embryo at five different stages between 6 and 96 hpf (hours post-fertilization). Biocompatibility and toxicity studies indicate that the NPs are not toxic and the embryos were found to exhibit normal morphological and gene expression in addition to the proper form and function of major organs such as the heart and vasculature (of 7 day old NPs injected zebrafish embryos). A multiplex in vivo detection protocol was developed by SERS imaging to demonstrate that multiple labels can be detected by Raman mapping in undifferentiated cells as they develop into distinct cell- and tissue-types. The present work is the first to report on multiplex Raman imaging of zebrafish embryos with potential implications in tracking tissue development and biological processes at single molecule sensitivity using appropriate target molecules in vivo.
Collapse
Affiliation(s)
- Yuling Wang
- Bindley Biosciences Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|
42
|
Verma MK, Lenka N. Temporal and contextual orchestration of cardiac fate by WNT-BMP synergy and threshold. J Cell Mol Med 2010; 14:2094-108. [PMID: 19432819 PMCID: PMC3823001 DOI: 10.1111/j.1582-4934.2009.00774.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cardiomyogenic development proceeds with a cascade of intricate signalling events that operate in a temporo-spatial fashion to specify cardiac cell fate during early embryogenesis. In fact, conflicting reports exist regarding the role of Wnt/β-catenin signalling during cardiomyogenesis. Here, we describe a dose-dependent and temporal effect of Wnt/β-catenin signalling on in vitro cardiomyogenesis using embryonic stem cells (ESCs) as a model system. We could demonstrate that canonical Wnt activation during early stage of differentiation either through ligand or by GSK3β inhibition helped in maintaining Oct4 and Nanog expressions, and in parallel, it promoted mesoderm and endoderm inductions. In contrast, it led to attenuation in cardiomyogenesis that was reversed by moderate concentration of DKK1, but not soluble Fz8. However, higher DKK1 could also block cardiomyogenesis, suggesting thereby governance of a particular signalling threshold underlying this developmental event. Interestingly, Wnt signalling activation at early stage modulated BMP4 expression in a stage-specific manner. Wnt activation, synchronized with BMP4 and brachyury up-regulation at early stage, correlated well with mesoderm induction. Conversely, Wnt activation led to BMP4 and Wnt5a down-regulation at late stage culminating in cardiomyogenic attenuation. Our findings suggested the existence of precise regulatory machinery with context-dependent role of Wnt for fine tuning mesoderm induction and its derivatives, through establishment of Wnt gradient during ESCs’ differentiation. Moreover, contrary to mere activation/inhibition, a specific threshold of Wnt and BMP and their synergy seemed necessary for providing the guiding cues in orchestrating mesoderm induction and subsequent cardiomyogenesis.
Collapse
Affiliation(s)
- Mahesh K Verma
- National Centre for Cell Science, Ganeshkhind, Pune, Maharashtra, India
| | | |
Collapse
|
43
|
Itasaki N, Hoppler S. Crosstalk between Wnt and bone morphogenic protein signaling: a turbulent relationship. Dev Dyn 2010; 239:16-33. [PMID: 19544585 DOI: 10.1002/dvdy.22009] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Wnt and the bone morphogenic protein (BMP) pathways are evolutionarily conserved and essentially independent signaling mechanisms, which, however, often regulate similar biological processes. Wnt and BMP signaling are functionally integrated in many biological processes, such as embryonic patterning in Drosophila and vertebrates, formation of kidney, limb, teeth and bones, maintenance of stem cells, and cancer progression. Detailed inspection of regulation in these and other tissues reveals that Wnt and BMP signaling are functionally integrated in four fundamentally different ways. The molecular mechanism evolved to mediate this integration can also be summarized in four different ways. However, a fundamental aspect of functional and mechanistic interaction between these pathways relies on tissue-specific mechanisms, which are often not conserved and cannot be extrapolated to other tissues. Integration of the two pathways contributes toward the sophisticated means necessary for creating the complexity of our bodies and the reliable and healthy function of its tissues and organs.
Collapse
Affiliation(s)
- Nobue Itasaki
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom.
| | | |
Collapse
|
44
|
Bai T, Seebald JL, Kim KE, Ding HM, Szeto DP, Chang HC. Disruption of zebrafish cyclin G-associated kinase (GAK) function impairs the expression of Notch-dependent genes during neurogenesis and causes defects in neuronal development. BMC DEVELOPMENTAL BIOLOGY 2010; 10:7. [PMID: 20082716 PMCID: PMC2821301 DOI: 10.1186/1471-213x-10-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 01/18/2010] [Indexed: 12/19/2022]
Abstract
Background The J-domain-containing protein auxilin, a critical regulator in clathrin-mediated transport, has been implicated in Drosophila Notch signaling. To ask if this role of auxilin is conserved and whether auxilin has additional roles in development, we have investigated the functions of auxilin orthologs in zebrafish. Results Like mammals, zebrafish has two distinct auxilin-like molecules, auxilin and cyclin G-associated kinase (GAK), differing in their domain structures and expression patterns. Both zebrafish auxilin and GAK can functionally substitute for the Drosophila auxilin, suggesting that they have overlapping molecular functions. Still, they are not completely redundant, as morpholino-mediated knockdown of the ubiquitously expressed GAK alone can increase the specification of neuronal cells, a known Notch-dependent process, and decrease the expression of Her4, a Notch target gene. Furthermore, inhibition of GAK function caused an elevated level of apoptosis in neural tissues, resulting in severe degeneration of neural structures. Conclusion In support of the notion that endocytosis plays important roles in Notch signaling, inhibition of zebrafish GAK function affects embryonic neuronal cell specification and Her4 expression. In addition, our analysis suggests that zebrafish GAK has at least two functions during the development of neural tissues: an early Notch-dependent role in neuronal patterning and a late role in maintaining the survival of neural cells.
Collapse
Affiliation(s)
- Ting Bai
- Department of Biological Sciences, Purdue University, 915 W, State St,, West Lafayette, Indiana 47907-2054, USA
| | | | | | | | | | | |
Collapse
|
45
|
Tanaka M, Jokubaitis V, Wood C, Wang Y, Brouard N, Pera M, Hearn M, Simmons P, Nakayama N. BMP inhibition stimulates WNT-dependent generation of chondrogenic mesoderm from embryonic stem cells. Stem Cell Res 2009; 3:126-41. [PMID: 19700382 DOI: 10.1016/j.scr.2009.07.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 06/22/2009] [Accepted: 07/10/2009] [Indexed: 10/20/2022] Open
Abstract
WNT and bone morphogenetic protein (BMP) signaling are known to stimulate hemogenesis from pluripotent embryonic stem (ES) cells. However, osteochondrogenic mesoderm was generated effectively when BMP signaling is kept to a low level, while WNT signaling was strongly activated. When mesoderm specification from ES cells was exogenous factor dependent, WNT3a addition supported the generation of cardiomyogenic cells expressing lateral plate/extraembryonic mesoderm genes, and this process involved endogenous BMP activities. Exogenous BMP4 showed a similar effect that depended on endogenous WNT activities. However, neither factor induced robust chondrogenic activity. In support, ES cell differentiation in the presence of either WNT3a or BMP4 was associated with elevated levels of both Bmp and Wnt mRNAs, which appeared to provide sufficient levels of active BMPs and WNTs to promote the nonchondrogenic mesoderm specification. The osteochondrogenic mesoderm expressed PDGFRalpha, which also expressed genes that mark somite and rostral presomitic mesoderm. A strong WNT signaling was required for generating the mesodermal progeny, while approximately 50- to 100-fold lower concentration of WNT3a was sufficient for specifying axial mes(end)oderm. Thus, depending on the dose and cofactor (BMP), WNT signaling stimulates the generation of different biological activities and specification of different types of mesodermal progeny from ES cells.
Collapse
Affiliation(s)
- Makoto Tanaka
- Peter MacCallum Cancer Institute, East Melbourne VIC 3002, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Chen YL, Liu B, Zhou ZN, Hu RY, Fei C, Xie ZH, Ding X. Smad6 inhibits the transcriptional activity of Tbx6 by mediating its degradation. J Biol Chem 2009; 284:23481-90. [PMID: 19561075 DOI: 10.1074/jbc.m109.007864] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Members of the bone morphogenetic protein (BMP) and T-box gene families play several critical roles in the early embryonic development and tissue homeostasis. Although BMP proteins are the upstream regulators of T-box genes, few studies have investigated the molecular mechanisms between these two protein families. Here, we report that Tbx6 interacts directly with Smad6, an inhibitory Smad that antagonizes the BMP signal. This interaction is mediated through the Mad homology 2 (MH2) domain of Smad6 and residues 90-180 of Tbx6. We demonstrate that Smad6 facilitates the degradation of Tbx6 protein through recruitment of Smurf1, a ubiquitin E3 ligase. Consequently, Smad6 reduces Tbx6-mediated Myf-5 gene activation. Furthermore, specific knockdown of endogenous Smad6 and Smurf1 by small interfering RNA increases the protein levels of Tbx6 and enhance the expression of Tbx6 target genes. Collectively, these findings reveal that Smad6 serves as a critical mediator of BMP signal via a functional interaction with Tbx6, thus regulating the activation of Tbx6 downstream genes during cell differentiation.
Collapse
Affiliation(s)
- Yue-Lei Chen
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | |
Collapse
|
47
|
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: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
48
|
Row RH, Kimelman D. Bmp inhibition is necessary for post-gastrulation patterning and morphogenesis of the zebrafish tailbud. Dev Biol 2009; 329:55-63. [PMID: 19236859 DOI: 10.1016/j.ydbio.2009.02.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 02/08/2009] [Accepted: 02/11/2009] [Indexed: 01/20/2023]
Abstract
Intricate interactions between the Wnt and Bmp signaling pathways pattern the gastrulating vertebrate embryo using a network of secreted protein ligands and inhibitors. While many of these proteins are expressed post-gastrula, their later roles have typically remained unclear, obscured by the effects of early perturbation. We find that Bmp signaling continues during somitogenesis in zebrafish embryos, with high activity in a small region of the mesodermal progenitor zone at the posterior end of the embryo. To test the hypothesis that Bmp inhibitors expressed just anterior to the tailbud are important to restrain Bmp signaling we produced a new zebrafish transgenic line, allowing temporal cell-autonomous activation of Bmp signaling and thereby bypassing the effects of the Bmp inhibitors. Ectopic activation of Bmp signaling during somitogenesis results in severe defects in the tailbud, including altered morphogenesis and gene expression. We show that these defects are due to non-autonomous effects on the tailbud, and present evidence that the tailbud defects are caused by alterations in Wnt signaling. We present a model in which the posteriorly expressed Bmp inhibitors function during somitogenesis to constrain Bmp signaling in the tailbud in order to allow normal expression of Wnt inhibitors in the presomitic mesoderm, which in turn constrain the levels of canonical and non-canonical Wnt signaling in the tailbud.
Collapse
Affiliation(s)
- Richard H Row
- Department of Biochemistry, University of Washington, Seattle, WA 98195-7350, USA
| | | |
Collapse
|
49
|
Lengerke C, Schmitt S, Bowman TV, Jang IH, Maouche-Chretien L, McKinney-Freeman S, Davidson AJ, Hammerschmidt M, Rentzsch F, Green JBA, Zon LI, Daley GQ. BMP and Wnt specify hematopoietic fate by activation of the Cdx-Hox pathway. Cell Stem Cell 2009; 2:72-82. [PMID: 18371423 DOI: 10.1016/j.stem.2007.10.022] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 10/11/2007] [Accepted: 10/31/2007] [Indexed: 01/10/2023]
Abstract
The formation of blood in the embryo is dependent on bone morphogenetic protein (BMP), but how BMP signaling intersects with other regulators of hematopoietic development is unclear. Using embryonic stem (ES) cells, we show that BMP4 first induces ventral-posterior (V-P) mesoderm and subsequently directs mesodermal cells toward blood fate by activating Wnt3a and upregulating Cdx and Hox genes. When BMP signaling is blocked during this latter phase, enforced expression of either Cdx1 or Cdx4 rescues hematopoietic development, thereby placing BMP4 signaling upstream of the Cdx-Hox pathway. Wnt signaling cooperates in BMP-induced hemogenesis, and the Wnt effector LEF1 mediates BMP4 activation of Cdx genes. Our data suggest that BMP signaling plays two distinct and sequential roles during blood formation, initially as an inducer of mesoderm, and later to specify blood via activation of Wnt signaling and the Cdx-Hox pathway.
Collapse
Affiliation(s)
- Claudia Lengerke
- Division of Pediatric Hematology/Oncology, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Veien ES, Rosenthal JS, Kruse-Bend RC, Chien CB, Dorsky RI. Canonical Wnt signaling is required for the maintenance of dorsal retinal identity. Development 2008; 135:4101-11. [PMID: 19004855 DOI: 10.1242/dev.027367] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Accurate retinotectal axon pathfinding depends upon the correct establishment of dorsal-ventral retinal polarity. We show that dorsal retinal gene expression is regulated by Wnt signaling in the dorsal retinal pigment epithelium (RPE). We find that a Wnt reporter transgene and Wnt pathway components are expressed in the dorsal RPE beginning at 14-16 hours post-fertilization. In the absence of Wnt signaling, tbx5 and Bmp genes initiate normal dorsal retinal expression but are not maintained. The expression of these genes is rescued by the downstream activation of Wnt signaling, and tbx5 is rescued by Bmp signaling. Furthermore, activation of Wnt signaling cannot rescue tbx5 in the absence of Bmp signaling, suggesting that Wnt signaling maintains dorsal retinal gene expression by regulating Bmp signaling. We present a model in which dorsal RPE-derived Wnt activity maintains the expression of Bmp ligands in the dorsal retina, thus coordinating the patterning of these two ocular tissues.
Collapse
Affiliation(s)
- Eric S Veien
- Department of Neurobiology and Anatomy, University of Utah, 401 MREB, 20 N. 1900 E., Salt Lake City, UT 84132, USA
| | | | | | | | | |
Collapse
|