1
|
He Z, Chen Q, Xiong J, Chen M, Gao K, Lai B, Ding W, Huang J, Zheng L, Pu Y, Tang Z, Zhang M, Yang D, Yan T. FoxH1 Represses the Promoter Activity of cyp19a1a in the Ricefield Eel ( Monopterus albus). Int J Mol Sci 2023; 24:13712. [PMID: 37762014 PMCID: PMC10531137 DOI: 10.3390/ijms241813712] [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: 06/30/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Forkhead box H1 (FoxH1) is a sexually dimorphic gene in Oreochromis niloticus, Oplegnathus fasciatus, and Acanthopagrus latus, indicating that it is essential for gonadal development. In the present study, the molecular characteristics and potential function of FoxH1 and the activation of the cyp19a1a promoter in vitro were evaluated in Monopterus albus. The levels of foxh1 in the ovaries were three times higher than those in the testes and were regulated by gonadotropins (Follicle-Stimulating Hormone and Human Chorionic Gonadotropin). FoxH1 colocalized with Cyp19a1a in the oocytes and granulosa cells of middle and late vitellogenic follicles. In addition, three FoxH1 binding sites were identified in the proximal promoter of cyp19a1a, namely, FH1 (-871/-860), FH2 (-535/-524), and FH3 (-218/-207). FoxH1 overexpression significantly attenuated the activity of the cyp19a1a promoter in CHO cells, and FH1/2 mutation increased promoter activity. Taken together, these results suggest that FoxH1 may act as an important regulator in the ovarian development of M. albus by repressing cyp19a1a promoter activity, which provides a foundation for the study of FoxH1 function in bony fish reproductive processes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Deying Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Z.H.); (Q.C.)
| | - Taiming Yan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Z.H.); (Q.C.)
| |
Collapse
|
2
|
Keum BR, Yeo I, Koo Y, Han W, Choi SC, Kim GH, Han JK. Transmembrane protein 150b attenuates BMP signaling in the Xenopus organizer. J Cell Physiol 2023; 238:1850-1866. [PMID: 37435758 DOI: 10.1002/jcp.31059] [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: 10/27/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 07/13/2023]
Abstract
The vertebrate organizer is a specified embryonic tissue that regulates dorsoventral patterning and axis formation. Although numerous cellular signaling pathways have been identified as regulators of the organizer's dynamic functions, the process remains incompletely understood, and as-yet unknown pathways remain to be explored for sophisticated mechanistic understanding of the vertebrate organizer. To identify new potential key factors of the organizer, we performed complementary DNA (cDNA) microarray screening using organizer-mimicking Xenopus laevis tissue. This analysis yielded a list of prospective organizer genes, and we determined the role of six-transmembrane domain containing transmembrane protein 150b (Tmem150b) in organizer function. Tmem150b was expressed in the organizer region and induced by Activin/Nodal signaling. In X. laevis, Tmem150b knockdown resulted in head defects and a shortened body axis. Moreover, Tmem150b negatively regulated bone morphogenetic protein (BMP) signaling, likely via physical interaction with activin receptor-like kinase 2 (ALK2). These findings demonstrated that Tmem150b functions as a novel membrane regulatory factor of BMP signaling with antagonistic effects, contributing to the understanding of regulatory molecular mechanisms of organizer axis function. Investigation of additional candidate genes identified in the cDNA microarray analysis could further delineate the genetic networks of the organizer during vertebrate embryogenesis.
Collapse
Affiliation(s)
- Byeong-Rak Keum
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
- Research Center for drug development, CYPHARMA, Daejeon, Korea
| | - Inchul Yeo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Youngmu Koo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Wonhee Han
- Department of Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Gun-Hwa Kim
- Research Center for drug development, CYPHARMA, Daejeon, Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Korea
| | - Jin-Kwan Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| |
Collapse
|
3
|
Favarolo MB, Revinski DR, Garavaglia MJ, López SL. Nodal and churchill1 position the expression of a notch ligand during Xenopus germ layer segregation. Life Sci Alliance 2022; 5:5/12/e202201693. [PMID: 36180230 PMCID: PMC9604498 DOI: 10.26508/lsa.202201693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Churchill and Nodal signaling, which participate in vertebrates’ germ layer induction, position a domain of Delta/Notch activity, which refines germ layer boundaries during frog gastrulation. In vertebrates, Nodal signaling plays a major role in endomesoderm induction, but germ layer delimitation is poorly understood. In avian embryos, the neural/mesoderm boundary is controlled by the transcription factor CHURCHILL1, presumably through the repressor ZEB2, but there is scarce knowledge about its role in other vertebrates. During amphibian gastrulation, Delta/Notch signaling refines germ layer boundaries in the marginal zone, but it is unknown the place this pathway occupies in the network comprising Churchill1 and Nodal. Here, we show that Xenopus churchill1 is expressed in the presumptive neuroectoderm at mid-blastula transition and during gastrulation, upregulates zeb2, prevents dll1 expression in the neuroectoderm, and favors neuroectoderm over endomesoderm development. Nodal signaling prevents dll1 expression in the endoderm but induces it in the presumptive mesoderm, from where it activates Notch1 and its target gene hes4 in the non-involuting marginal zone. We propose a model where Nodal and Churchill1 position Dll1/Notch1/Hes4 domains in the marginal zone, ensuring the delimitation between mesoderm and neuroectoderm.
Collapse
Affiliation(s)
- María Belén Favarolo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología/1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| | - Diego R Revinski
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología/1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| | - Matías J Garavaglia
- Laboratorio de Bioinsumos, Instituto de Biotecnología, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología/1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina .,CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| |
Collapse
|
4
|
Seudre O, Martín-Zamora FM, Rapisarda V, Luqman I, Carrillo-Baltodano AM, Martín-Durán JM. The Fox Gene Repertoire in the Annelid Owenia fusiformis Reveals Multiple Expansions of the foxQ2 Class in Spiralia. Genome Biol Evol 2022; 14:evac139. [PMID: 36099507 PMCID: PMC9539403 DOI: 10.1093/gbe/evac139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
Fox genes are a large and conserved family of transcription factors involved in many key biological processes, including embryogenesis and body patterning. Although the role of Fox genes has been studied in an array of model systems, comprehensive comparative studies in Spiralia-a large clade of invertebrate animals including molluscs and annelids-are scarce but much needed to better understand the evolutionary history of this gene family. Here, we reconstruct and functionally characterize the Fox gene complement in the annelid Owenia fusiformis, a slow evolving species and member of the sister group to all remaining annelids. The genome of O. fusiformis contains at least a single ortholog for 20 of the 22 Fox gene classes that are ancestral to Bilateria, including an ortholog of the recently discovered foxT class. Temporal and spatial expression dynamics reveal a conserved role of Fox genes in gut formation, mesoderm patterning, and apical organ and cilia formation in Annelida and Spiralia. Moreover, we uncover an ancestral expansion of foxQ2 genes in Spiralia, represented by 11 paralogs in O. fusiformis. Notably, although all foxQ2 copies have apical expression in O. fusiformis, they show variable spatial domains and staggered temporal activation, which suggest cooperation and sub-functionalization among foxQ2 genes for the development of apical fates in this annelid. Altogether, our study informs the evolution and developmental roles of Fox genes in Annelida and Spiralia generally, providing the basis to explore how regulatory changes in Fox gene expression might have contributed to developmental and morphological diversification in Spiralia.
Collapse
Affiliation(s)
- Océane Seudre
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| | - Francisco M Martín-Zamora
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| | - Valentina Rapisarda
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| | - Imran Luqman
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| | - Allan M Carrillo-Baltodano
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| | - José M Martín-Durán
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, E1 4NSUnited Kingdom
| |
Collapse
|
5
|
Zhou JJ, Pham PD, Han H, Wang W, Cho KW. Engagement of Foxh1 in chromatin regulation revealed by protein interactome analyses. Dev Growth Differ 2022; 64:297-305. [PMID: 35848281 PMCID: PMC9474667 DOI: 10.1111/dgd.12799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022]
Abstract
Early embryonic cell fates are specified through coordinated integration of transcription factor activities and epigenetic states of the genome. Foxh1 is a key maternal transcription factor controlling the mesendodermal gene regulatory program. Proteomic interactome analyses using FOXH1 as a bait in mouse embryonic stem cells revealed that FOXH1 interacts with PRC2 subunits and HDAC1. Foxh1 physically interacts with Hdac1, and confers transcriptional repression of mesendodermal genes in Xenopus ectoderm. Our findings reveal a central role of Foxh1 in coordinating the chromatin states of the Xenopus embryonic genome.
Collapse
Affiliation(s)
- Jeff Jiajing Zhou
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Paula Duyen Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Han Han
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Ken W.Y. Cho
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- Lead Contact
| |
Collapse
|
6
|
Tao W, Shi H, Yang J, Diakite H, Kocher TD, Wang D. Homozygous mutation of foxh1 arrests oogenesis causing infertility in female Nile tilapia†. Biol Reprod 2021; 102:758-769. [PMID: 31837141 DOI: 10.1093/biolre/ioz225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/13/2019] [Accepted: 12/13/2019] [Indexed: 01/15/2023] Open
Abstract
Foxh1, a member of fox gene family, was first characterized as a transcriptional partner in the formation of the Smad protein complex. Recent studies have shown foxh1 is highly expressed in the cytoplasm of oocytes in both tilapia and mouse. However, its function in oogenesis remains unexplored. In the present study, foxh1-/- tilapia was created by CRISPR/Cas9. At 180 dah (days after hatching), the foxh1-/- XX fish showed oogenesis arrest and a significantly lower GSI. The transition of oocytes from phase II to phase III and follicle cells from one to two layers was blocked, resulting in infertility of the mutant. Transcriptomic analysis revealed that expression of genes involved in estrogen synthesis and oocyte growth were altered in the foxh1-/- ovaries. Loss of foxh1 resulted in significantly decreased Cyp19a1a and increased Cyp11b2 expression, consistent with significantly lower concentrations of serum estradiol-17β (E2) and higher concentrations of 11-ketotestosterone (11-KT). Moreover, administration of E2 rescued the phenotypes of foxh1-/- XX fish, as indicated by the appearance of phase III and IV oocytes and absence of Cyp11b2 expression. Taken together, these results suggest that foxh1 functions in the oocytes to regulate oogenesis by promoting cyp19a1a expression, and therefore estrogen production. Disruption of foxh1 may block the estrogen synthesis and oocyte growth, leading to the arrest of oogenesis and thus infertility in tilapia.
Collapse
Affiliation(s)
- Wenjing Tao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hongjuan Shi
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.,Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China and
| | - Jing Yang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hamidou Diakite
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Thomas D Kocher
- Department of Biology, University of Maryland, College Park, Maryland, United States of America
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
7
|
Fuentes R, Tajer B, Kobayashi M, Pelliccia JL, Langdon Y, Abrams EW, Mullins MC. The maternal coordinate system: Molecular-genetics of embryonic axis formation and patterning in the zebrafish. Curr Top Dev Biol 2020; 140:341-389. [PMID: 32591080 DOI: 10.1016/bs.ctdb.2020.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Axis specification of the zebrafish embryo begins during oogenesis and relies on proper formation of well-defined cytoplasmic domains within the oocyte. Upon fertilization, maternally-regulated cytoplasmic flow and repositioning of dorsal determinants establish the coordinate system that will build the structure and developmental body plan of the embryo. Failure of specific genes that regulate the embryonic coordinate system leads to catastrophic loss of body structures. Here, we review the genetic principles of axis formation and discuss how maternal factors orchestrate axis patterning during zebrafish early embryogenesis. We focus on the molecular identity and functional contribution of genes controlling critical aspects of oogenesis, egg activation, blastula, and gastrula stages. We examine how polarized cytoplasmic domains form in the oocyte, which set off downstream events such as animal-vegetal polarity and germ line development. After gametes interact and form the zygote, cytoplasmic segregation drives the animal-directed reorganization of maternal determinants through calcium- and cell cycle-dependent signals. We also summarize how maternal genes control dorsoventral, anterior-posterior, mesendodermal, and left-right cell fate specification and how signaling pathways pattern these axes and tissues during early development to instruct the three-dimensional body plan. Advances in reverse genetics and phenotyping approaches in the zebrafish model are revealing positional patterning signatures at the single-cell level, thus enhancing our understanding of genotype-phenotype interactions in axis formation. Our emphasis is on the genetic interrogation of novel and specific maternal regulatory mechanisms of axis specification in the zebrafish.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Manami Kobayashi
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Jose L Pelliccia
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | | | - Elliott W Abrams
- Department of Biology, Purchase College, State University of New York, Harrison, NY, United States
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
8
|
Paraiso KD, Cho JS, Yong J, Cho KWY. Early Xenopus gene regulatory programs, chromatin states, and the role of maternal transcription factors. Curr Top Dev Biol 2020; 139:35-60. [PMID: 32450966 PMCID: PMC11344482 DOI: 10.1016/bs.ctdb.2020.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
For decades, the early development of the Xenopus embryo has been an essential model system to study the gene regulatory mechanisms that govern cellular specification. At the top of the hierarchy of gene regulatory networks, maternally deposited transcription factors initiate this process and regulate the expression of zygotic genes that give rise to three distinctive germ layer cell types (ectoderm, mesoderm, and endoderm), and subsequent generation of organ precursors. The onset of germ layer specification is also closely coupled with changes associated with chromatin modifications. This review will examine the timing of maternal transcription factors initiating the zygotic genome activation, the epigenetic landscape of embryonic chromatin, and the network structure that governs the process.
Collapse
Affiliation(s)
- Kitt D Paraiso
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States; Center for Complex Biological Systems, University of California, Irvine, CA, United States
| | - Jin S Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Junseok Yong
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States; Center for Complex Biological Systems, University of California, Irvine, CA, United States.
| |
Collapse
|
9
|
Fischer P, Chen H, Pacho F, Rieder D, Kimmel RA, Meyer D. FoxH1 represses miR-430 during early embryonic development of zebrafish via non-canonical regulation. BMC Biol 2019; 17:61. [PMID: 31362746 PMCID: PMC6664792 DOI: 10.1186/s12915-019-0683-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND FoxH1 is a forkhead transcription factor with conserved key functions in vertebrate mesoderm induction and left-right patterning downstream of the TGF-beta/Nodal signaling pathway. Binding of the forkhead domain (FHD) of FoxH1 to a highly conserved proximal sequence motif was shown to regulate target gene expression. RESULTS We identify the conserved microRNA-430 family (miR-430) as a novel target of FoxH1. miR-430 levels are increased in foxH1 mutants, resulting in a reduced expression of transcripts that are targeted by miR-430 for degradation. To determine the underlying mechanism of miR-430 repression, we performed chromatin immunoprecipitation studies and overexpression experiments with mutant as well as constitutive active and repressive forms of FoxH1. Our studies reveal a molecular interaction of FoxH1 with miR-430 loci independent of the FHD. Furthermore, we show that previously described mutant forms of FoxH1 that disrupt DNA binding or that lack the C-terminal Smad Interaction Domain (SID) dominantly interfere with miR-430 repression, but not with the regulation of previously described FoxH1 targets. CONCLUSIONS We were able to identify the distinct roles of protein domains of FoxH1 in the regulation process of miR-430. We provide evidence that the indirect repression of miR-430 loci depends on the connection to a distal repressive chromosome environment via a non-canonical mode. The widespread distribution of such non-canonical binding sites of FoxH1, found not only in our study, argues against a function restricted to regulating miR-430 and for a more global role of FoxH1 in chromatin folding.
Collapse
Affiliation(s)
- Patrick Fischer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Hao Chen
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Frederic Pacho
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Dietmar Rieder
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innrain 80, 6020, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria.
| |
Collapse
|
10
|
Sena E, Rocques N, Borday C, Amin HSM, Parain K, Sitbon D, Chesneau A, Durand BC. Barhl2 maintains T-cell factors as repressors, and thereby switches off the Wnt/β-Catenin response driving Spemann organizer formation. Development 2019; 146:dev.173112. [DOI: 10.1242/dev.173112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/11/2019] [Indexed: 12/19/2022]
Abstract
A hallmark of Wnt/β-Catenin signaling is the extreme diversity of its transcriptional response, which varies depending on cell and developmental context. What controls this diversity is poorly understood. In all cases, the switch from transcriptional repression to activation depends on a nuclear increase in β-Catenin, which detaches the transcription factor T-cell Factor-7 like 1 (Tcf7l1) bound to Groucho (Gro) transcriptional co-repressors from its DNA binding sites and transiently converts Tcf7/Lymphoid enhancer binding factor 1 (Lef1) into a transcriptional activator. One of the earliest and evolutionarily conserved functions of Wnt/β-Catenin signaling is the induction of the blastopore lip organizer. Here, we demonstrate that the evolutionarily conserved BarH-like homeobox-2 (Barhl2) protein stabilizes the Tcf7l1-Gro complex and maintains repressed expression of Tcf target genes by a mechanism that depends on histone deacetylase 1 (Hdac-1) activity. In this way, Barhl2 switches off the Wnt/β-Catenin-dependent early transcriptional response, thereby limiting the formation of the organizer in time and/or space. This study reveals a novel nuclear inhibitory mechanism of Wnt/Tcf signaling that switches off organizer fate determination.
Collapse
Affiliation(s)
- Elena Sena
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Nathalie Rocques
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
| | - Caroline Borday
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Harem Sabr Muhamad Amin
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| | - Karine Parain
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - David Sitbon
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Albert Chesneau
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Béatrice C. Durand
- Institut Curie, Research Division, PSL Research University, Université Paris Sud, CNRS UMR 3347, INSERM U1021, Centre Universitaire, Bâtiment 110 F-91405 Orsay Cedex
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 CNRS 8197, INSERM U1024 46 rue d'Ulm 75005, Paris F-75005, France
| |
Collapse
|
11
|
Cheng Q, Shi YJ, Li Z, Kang H, Xiang Z, Kong LF. FAST1 promotes the migration and invasion of colorectal cancer cells. Biochem Biophys Res Commun 2018; 509:407-413. [PMID: 30594391 DOI: 10.1016/j.bbrc.2018.12.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/16/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND The forkhead activin signal transducer 1 (FAST1) is involved in several oncogenic signaling pathways and its abnormal expression has been discovered in some cancers. Yet the role of FAST1 in colorectal cancer (CRC) remains largely unclear. Therefore, the goal of this study was to explore the function of FAST1 in CRC. METHODS In this study, we analyzed FAST1 expression and its relationship with clinicopathological parameters and prognostic significance in CRC via immunohistochemistry analysis. The effects and mechanisms of FAST1 on cell proliferation, migration and invasion were explored in vitro and in vivo. RESULTS We found that increased FAST1 as an independent prognostic factor was positively associated with TNM stage and pathological grade in CRC. FAST1 overexpression promoted the CRC cell proliferation, migration and invasion in vivo. Furthermore, mechanistic studies implicated that FAST1 enhanced the pulmonary metastasis of CRC cells through down-regulating E-cadherin levels. CONCLUSIONS In summary, FAST1 was significantly associated with CRC progression and could serve as an independent prognostic factor. FAST1 may be potential therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Qiong Cheng
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Yu-Jie Shi
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Zhen Li
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Hong Kang
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Zheng Xiang
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Ling-Fei Kong
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
12
|
Gere-Becker MB, Pommerenke C, Lingner T, Pieler T. Retinoic acid-induced expression of Hnf1b and Fzd4 is required for pancreas development in Xenopus laevis. Development 2018; 145:dev.161372. [PMID: 29769220 PMCID: PMC6031401 DOI: 10.1242/dev.161372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 05/04/2018] [Indexed: 12/17/2022]
Abstract
Retinoic acid (RA) is required for pancreas specification in Xenopus and other vertebrates. However, the gene network that is directly induced by RA signalling in this context remains to be defined. By RNA sequencing of in vitro-generated pancreatic explants, we identified the genes encoding the transcription factor Hnf1β and the Wnt-receptor Fzd4/Fzd4s as direct RA target genes. Functional analyses of Hnf1b and Fzd4/Fzd4s in programmed pancreatic explants and whole embryos revealed their requirement for pancreatic progenitor formation and differentiation. Thus, Hnf1β and Fzd4/Fzd4s appear to be involved in pre-patterning events of the embryonic endoderm that allow pancreas formation in Xenopus.
Collapse
Affiliation(s)
- Maja B Gere-Becker
- Department of Developmental Biochemistry, University of Goettingen, Justus-von-Liebig-Weg 11, 37077 Goettingen, Germany
| | - Claudia Pommerenke
- Department of Developmental Biochemistry, University of Goettingen, Justus-von-Liebig-Weg 11, 37077 Goettingen, Germany.,Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Inhoffenstrasse 7B, 38124 Braunschweig, Germany
| | - Thomas Lingner
- Department of Developmental Biochemistry, University of Goettingen, Justus-von-Liebig-Weg 11, 37077 Goettingen, Germany.,Genevention GmbH, Rudolf-Wissel-Str. 28, 37079 Goettingen, Germany
| | - Tomas Pieler
- Department of Developmental Biochemistry, University of Goettingen, Justus-von-Liebig-Weg 11, 37077 Goettingen, Germany
| |
Collapse
|
13
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
14
|
Tadjuidje E, Kofron M, Mir A, Wylie C, Heasman J, Cha SW. Nodal signalling in Xenopus: the role of Xnr5 in left/right asymmetry and heart development. Open Biol 2017; 6:rsob.150187. [PMID: 27488374 PMCID: PMC5008007 DOI: 10.1098/rsob.150187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 07/01/2016] [Indexed: 01/05/2023] Open
Abstract
Nodal class TGF-β signalling molecules play essential roles in establishing the vertebrate body plan. In all vertebrates, nodal family members have specific waves of expression required for tissue specification and axis formation. In Xenopus laevis, six nodal genes are expressed before gastrulation, raising the question of whether they have specific roles or act redundantly with each other. Here, we examine the role of Xnr5. We find it acts at the late blastula stage as a mesoderm inducer and repressor of ectodermal gene expression, a role it shares with Vg1. However, unlike Vg1, Xnr5 depletion reduces the expression of the nodal family member xnr1 at the gastrula stage. It is also required for left/right laterality by controlling the expression of the laterality genes xnr1, antivin (lefty) and pitx2 at the tailbud stage. In Xnr5-depleted embryos, the heart field is established normally, but symmetrical reduction in Xnr5 levels causes a severely stunted midline heart, first evidenced by a reduction in cardiac troponin mRNA levels, while left-sided reduction leads to randomization of the left/right axis. This work identifies Xnr5 as the earliest step in the signalling pathway establishing normal heart laterality in Xenopus.
Collapse
Affiliation(s)
- Emmanuel Tadjuidje
- Department of Biological Sciences, Alabama State University, 1627 Hall Street, Montgomery, AL 36101, USA
| | - Matthew Kofron
- Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Adnan Mir
- Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Christopher Wylie
- Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Janet Heasman
- Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Sang-Wook Cha
- Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
15
|
Charney RM, Forouzmand E, Cho JS, Cheung J, Paraiso KD, Yasuoka Y, Takahashi S, Taira M, Blitz IL, Xie X, Cho KWY. Foxh1 Occupies cis-Regulatory Modules Prior to Dynamic Transcription Factor Interactions Controlling the Mesendoderm Gene Program. Dev Cell 2017; 40:595-607.e4. [PMID: 28325473 PMCID: PMC5434453 DOI: 10.1016/j.devcel.2017.02.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/24/2016] [Accepted: 02/16/2017] [Indexed: 12/14/2022]
Abstract
The interplay between transcription factors and chromatin dictates gene regulatory network activity. Germ layer specification is tightly coupled with zygotic gene activation and, in most metazoans, is dependent upon maternal factors. We explore the dynamic genome-wide interactions of Foxh1, a maternal transcription factor that mediates Nodal/TGF-β signaling, with cis-regulatory modules (CRMs) during mesendodermal specification. Foxh1 marks CRMs during cleavage stages and recruits the co-repressor Tle/Groucho in the early blastula. We highlight a population of CRMs that are continuously occupied by Foxh1 and show that they are marked by H3K4me1, Ep300, and Fox/Sox/Smad motifs, suggesting interplay between these factors in gene regulation. We also propose a molecular "hand-off" between maternal Foxh1 and zygotic Foxa at these CRMs to maintain enhancer activation. Our findings suggest that Foxh1 functions at the top of a hierarchy of interactions by marking developmental genes for activation, beginning with the onset of zygotic gene expression.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Elmira Forouzmand
- Department of Computer Science, Donald Bren School of Information & Computer Sciences, University of California, Irvine, CA 92697, USA
| | - Jin Sun Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Jessica Cheung
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Yuuri Yasuoka
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Shuji Takahashi
- Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8526, Japan
| | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Xiaohui Xie
- Department of Computer Science, Donald Bren School of Information & Computer Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
16
|
Charney RM, Paraiso KD, Blitz IL, Cho KWY. A gene regulatory program controlling early Xenopus mesendoderm formation: Network conservation and motifs. Semin Cell Dev Biol 2017; 66:12-24. [PMID: 28341363 DOI: 10.1016/j.semcdb.2017.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/12/2017] [Accepted: 03/20/2017] [Indexed: 02/08/2023]
Abstract
Germ layer formation is among the earliest differentiation events in metazoan embryos. In triploblasts, three germ layers are formed, among which the endoderm gives rise to the epithelial lining of the gut tube and associated organs including the liver, pancreas and lungs. In frogs (Xenopus), where early germ layer formation has been studied extensively, the process of endoderm specification involves the interplay of dozens of transcription factors. Here, we review the interactions between these factors, summarized in a transcriptional gene regulatory network (GRN). We highlight regulatory connections conserved between frog, fish, mouse, and human endodermal lineages. Especially prominent is the conserved role and regulatory targets of the Nodal signaling pathway and the T-box transcription factors, Vegt and Eomes. Additionally, we highlight network topologies and motifs, and speculate on their possible roles in development.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
17
|
Silva M, Vargas S, Coelho A, Dias A, Ferreira T, Morais A, Maia R, Kjöllerström P, Lavinha J, Faustino P. Hemorheological alterations in sickle cell anemia and their clinical consequences - The role of genetic modulators. Clin Hemorheol Microcirc 2017; 64:859-866. [PMID: 27814292 DOI: 10.3233/ch-168048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Sickle cell anemia (SCA) is an autosomal recessive disease caused by the HBB:c.20A>T mutation that leads to hemoglobin S synthesis. The disease presents with high clinical heterogeneity characterized by chronic hemolysis, recurrent episodes of vaso-oclusion and infection. This work aimed to characterize by in silico studies some genetic modulators of severe hemolysis and stroke risk in children with SCA, and understand their consequences at the hemorheological level.Association studies were performed between hemolysis biomarkers as well as the degree of cerebral vasculopathy and the inheritance of several polymorphic regions in genes related with vascular cell adhesion and vascular tonus in pediatric SCA patients. In silico tools (e.g. MatInspector) were applied to investigate the main variant consequences.Variants in vascular adhesion molecule-1 (VCAM1) gene promoter and endothelial nitric oxide synthase (NOS3) gene were significantly associated with higher degree of hemolysis and stroke events. They potentially modify transcription factor binding sites (e.g. VCAM1 rs1409419_T allele may lead to an EVI1 gain) or disturb the corresponding protein structure/function. Our findings emphasize the relevance of genetic variation in modulating the disease severity due to their effect on gene expression or modification of protein biological activities related with sickled erythrocyte/endothelial interactions and consequent hemorheological abnormalities.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal
| | - Sofia Vargas
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal
| | - Andreia Coelho
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal
| | - Alexandra Dias
- Departamento de Pediatria, Núcleo de Hematologia, Hospital Prof. Doutor Fernando Fonseca, Amadora, Portugal
| | - Teresa Ferreira
- Departamento de Pediatria, Núcleo de Hematologia, Hospital Prof. Doutor Fernando Fonseca, Amadora, Portugal
| | - Anabela Morais
- Departamento de Pediatria, Hospital de Santa Maria, CHLN, Lisboa, Portugal
| | - Raquel Maia
- Unidade de Hematologia, Hospital de Dona Estefânia, CHLC, Lisboa, Portugal
| | - Paula Kjöllerström
- Unidade de Hematologia, Hospital de Dona Estefânia, CHLC, Lisboa, Portugal
| | - João Lavinha
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal.,BioISI, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal.,Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
18
|
Sampath K, Robertson EJ. Keeping a lid on nodal: transcriptional and translational repression of nodal signalling. Open Biol 2016; 6:150200. [PMID: 26791244 PMCID: PMC4736825 DOI: 10.1098/rsob.150200] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nodal is an evolutionarily conserved member of the transforming growth factor-β (TGF-β) superfamily of secreted signalling factors. Nodal factors are known to play key roles in embryonic development and asymmetry in a variety of organisms ranging from hydra and sea urchins to fish, mice and humans. In addition to embryonic patterning, Nodal signalling is required for maintenance of human embryonic stem cell pluripotency and mis-regulated Nodal signalling has been found associated with tumour metastases. Therefore, precise and timely regulation of this pathway is essential. Here, we discuss recent evidence from sea urchins, frogs, fish, mice and humans that show a role for transcriptional and translational repression of Nodal signalling during early development.
Collapse
Affiliation(s)
- Karuna Sampath
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AJ, UK
| | | |
Collapse
|
19
|
Reid CD, Steiner AB, Yaklichkin S, Lu Q, Wang S, Hennessy M, Kessler DS. FoxH1 mediates a Grg4 and Smad2 dependent transcriptional switch in Nodal signaling during Xenopus mesoderm development. Dev Biol 2016; 414:34-44. [PMID: 27085753 DOI: 10.1016/j.ydbio.2016.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/28/2016] [Accepted: 04/06/2016] [Indexed: 02/07/2023]
Abstract
In the vertebrate blastula and gastrula the Nodal pathway is essential for formation of the primary germ layers and the organizer. Nodal autoregulatory feedback potentiates signaling activity, but mechanisms limiting embryonic Nodal ligand transcription are poorly understood. Here we describe a transcriptional switch mechanism mediated by FoxH1, the principle effector of Nodal autoregulation. FoxH1 contains a conserved engrailed homology (EH1) motif that mediates direct binding of groucho-related gene 4 (Grg4), a Groucho family corepressor. Nodal-dependent gene expression is suppressed by FoxH1, but enhanced by a FoxH1 EH1 mutant, indicating that the EH1 motif is necessary for repression. Grg4 blocks Nodal-induced mesodermal gene expression and Nodal autoregulation, suggesting that Grg4 limits Nodal pathway activity. Conversely, blocking Grg4 function in the ectoderm results in ectopic expression of Nodal target genes. FoxH1 and Grg4 occupy the Xnr1 enhancer, and Grg4 occupancy is dependent on the FoxH1 EH1 motif. Grg4 occupancy at the Xnr1 enhancer significantly decreases with Nodal activation or Smad2 overexpression, while FoxH1 occupancy is unaffected. These results suggest that Nodal-activated Smad2 physically displaces Grg4 from FoxH1, an essential feature of the transcriptional switch mechanism. In support of this model, when FoxH1 is unable to bind Smad2, Grg4 occupancy is maintained at the Xnr1 enhancer, even in the presence of Nodal signaling. Our findings reveal that FoxH1 mediates both activation and repression of Nodal gene expression. We propose that this transcriptional switch is essential to delimit Nodal pathway activity in vertebrate germ layer formation.
Collapse
Affiliation(s)
- Christine D Reid
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Aaron B Steiner
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Sergey Yaklichkin
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Qun Lu
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Shouwen Wang
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Morgan Hennessy
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| | - Daniel S Kessler
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine Smilow Center for Translational Research, Room 9-104, 3400 Civic Center Blvd Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Park S, Blaser S, Marchal MA, Houston DW, Sheets MD. A gradient of maternal Bicaudal-C controls vertebrate embryogenesis via translational repression of mRNAs encoding cell fate regulators. Development 2016; 143:864-71. [PMID: 26811381 PMCID: PMC4813341 DOI: 10.1242/dev.131359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/16/2016] [Indexed: 12/16/2022]
Abstract
Vertebrate Bicaudal-C (Bicc1) has important biological roles in the formation and homeostasis of multiple organs, but direct experiments to address the role of maternal Bicc1 in early vertebrate embryogenesis have not been reported. Here, we use antisense phosphorothioate-modified oligonucleotides and the host-transfer technique to eliminate specifically maternal stores of both bicc1 mRNA and Bicc1 protein from Xenopus laevis eggs. Fertilization of these Bicc1-depleted eggs produced embryos with an excess of dorsal-anterior structures and overexpressed organizer-specific genes, indicating that maternal Bicc1 is crucial for normal embryonic patterning of the vertebrate embryo. Bicc1 is an RNA-binding protein with robust translational repression function. Here, we show that the maternal mRNA encoding the cell-fate regulatory protein Wnt11b is a direct target of Bicc1-mediated repression. It is well established that the Wnt signaling pathway is crucial to vertebrate embryogenesis. Thus, the work presented here links the molecular function of Bicc1 in mRNA target-specific translation repression to its biological role in the maternally controlled stages of vertebrate embryogenesis.
Collapse
Affiliation(s)
- Sookhee Park
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Susanne Blaser
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | - Michael D Sheets
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
21
|
Chen L, Chu C, Kong X, Huang G, Huang T, Cai YD. A hybrid computational method for the discovery of novel reproduction-related genes. PLoS One 2015; 10:e0117090. [PMID: 25768094 PMCID: PMC4358884 DOI: 10.1371/journal.pone.0117090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 12/13/2014] [Indexed: 12/12/2022] Open
Abstract
Uncovering the molecular mechanisms underlying reproduction is of great importance to infertility treatment and to the generation of healthy offspring. In this study, we discovered novel reproduction-related genes with a hybrid computational method, integrating three different types of method, which offered new clues for further reproduction research. This method was first executed on a weighted graph, constructed based on known protein-protein interactions, to search the shortest paths connecting any two known reproduction-related genes. Genes occurring in these paths were deemed to have a special relationship with reproduction. These newly discovered genes were filtered with a randomization test. Then, the remaining genes were further selected according to their associations with known reproduction-related genes measured by protein-protein interaction score and alignment score obtained by BLAST. The in-depth analysis of the high confidence novel reproduction genes revealed hidden mechanisms of reproduction and provided guidelines for further experimental validations.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People’s Republic of China
| | - Chen Chu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People’s Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
| | - Guohua Huang
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
- * E-mail: (TH); (YDC)
| | - Yu-Dong Cai
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
- * E-mail: (TH); (YDC)
| |
Collapse
|
22
|
Chiu WT, Charney Le R, Blitz IL, Fish MB, Li Y, Biesinger J, Xie X, Cho KWY. Genome-wide view of TGFβ/Foxh1 regulation of the early mesendoderm program. Development 2014; 141:4537-47. [PMID: 25359723 DOI: 10.1242/dev.107227] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nodal/TGFβ signaling regulates diverse biological responses. By combining RNA-seq on Foxh1 and Nodal signaling loss-of-function embryos with ChIP-seq of Foxh1 and Smad2/3, we report a comprehensive genome-wide interaction between Foxh1 and Smad2/3 in mediating Nodal signaling during vertebrate mesendoderm development. This study significantly increases the total number of Nodal target genes regulated by Foxh1 and Smad2/3, and reinforces the notion that Foxh1-Smad2/3-mediated Nodal signaling directly coordinates the expression of a cohort of genes involved in the control of gene transcription, signaling pathway modulation and tissue morphogenesis during gastrulation. We also show that Foxh1 may function independently of Nodal signaling, in addition to its role as a transcription factor mediating Nodal signaling via Smad2/3. Finally, we propose an evolutionarily conserved interaction between Foxh1 and PouV, a mechanism observed in Pou5f1-mediated regulation of pluripotency in human embryonic stem and epiblast cells.
Collapse
Affiliation(s)
- William T Chiu
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Rebekah Charney Le
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Margaret B Fish
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Yi Li
- Department of Computer Science, University of California, Irvine, CA 92697-2300, USA
| | - Jacob Biesinger
- Department of Computer Science, University of California, Irvine, CA 92697-2300, USA
| | - Xiaohui Xie
- Department of Computer Science, University of California, Irvine, CA 92697-2300, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| |
Collapse
|
23
|
Gupta R, Wills A, Ucar D, Baker J. Developmental enhancers are marked independently of zygotic Nodal signals in Xenopus. Dev Biol 2014; 395:38-49. [PMID: 25205067 DOI: 10.1016/j.ydbio.2014.08.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/06/2014] [Accepted: 08/31/2014] [Indexed: 02/08/2023]
Abstract
To determine the hierarchy of transcriptional regulation within the in vivo vertebrate embryo, we examined whether developmental enhancers were influenced by Nodal signaling during early embryogenesis in Xenopus tropicalis. We find that developmental enhancers, defined by the active enhancer chromatin marks H3K4me1 and H3K27ac, are established as early as blastula stage and that Smad2/3 only strongly associates with these regions at gastrula stages. Significantly, when we perturb Nodal signaling using the drug SB431542, most enhancers remain marked, including at genes known to be sensitive to Nodal signaling. Overall, as enhancers are in an active conformation prior to Nodal signaling and are established independently of Nodal signaling, we suggest that many developmental enhancers are marked maternally, prior to exposure to extrinsic signals.
Collapse
Affiliation(s)
- Rakhi Gupta
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Andrea Wills
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Duygu Ucar
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Julie Baker
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.,Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Single blastomere expression profiling of Xenopus laevis embryos of 8 to 32-cells reveals developmental asymmetry. Sci Rep 2014; 3:2278. [PMID: 23880666 PMCID: PMC3721081 DOI: 10.1038/srep02278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/05/2013] [Indexed: 12/24/2022] Open
Abstract
We have measured the expression of 41 maternal mRNAs in individual blastomeres collected from the 8 to 32-cell Xenopus laevis embryos to determine when and how asymmetry in the body plan is introduced. We demonstrate that the asymmetry along the animal-vegetal axis in the oocyte is transferred to the daughter cells during early cell divisions. All studied mRNAs are distributed evenly among the set of animal as well as vegetal blastomeres. We find no asymmetry in mRNA levels that might be ascribed to the dorso-ventral specification or the left-right axis formation. We hypothesize that while the animal-vegetal asymmetry is a consequence of mRNA gradients, the dorso-ventral and left-right axes specifications are induced by asymmetric distribution of other biomolecules, probably proteins.
Collapse
|
25
|
Karadge U, Elinson RP. Characterization of the nutritional endoderm in the direct developing frog Eleutherodactylus coqui. Dev Genes Evol 2013; 223:351-62. [PMID: 24043604 DOI: 10.1007/s00427-013-0451-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/14/2013] [Indexed: 12/01/2022]
Abstract
Unlike Xenopus laevis, Eleutherodactylus coqui develops without a tadpole. The yolk-rich vegetal region of the embryo forms a transient nutritive tissue, the nutritional endoderm (NE). The definitive endoderm (DE) in E. coqui comes from cells closer to the animal pole in contrast to its vegetal origin in X. laevis. RNA important for initiating the endoderm specification network is absent in presumptive NE cells, raising the question whether signaling occurs in them. We explored the nature of NE and asked how differences between NE and DE cells arise. We identified differences between NE and DE that first become evident at gastrula, when NE cells become multinucleated. Nuclear β-catenin, an essential cofactor of sox 17, important for endoderm formation in X. laevis, is present in NE and DE at gastrula but remains in NE long after it is not seen in DE. We cloned E. coqui homologs of TGFβs activin b and derriere and provide evidence for their maternal expression. We also detected activin b and derriere RNAs in NE at gastrula and show that NE possesses some mesoderm-inducing activity, but it is delayed with respect to DE. Our findings indicate that altered development of NE begins at gastrula. RNAs important for mesendoderm induction and some mesoderm-inducing activity are present in NE.
Collapse
Affiliation(s)
- Uma Karadge
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA, USA,
| | | |
Collapse
|
26
|
Herriges JC, Yi L, Hines EA, Harvey JF, Xu G, Gray P, Ma Q, Sun X. Genome-scale study of transcription factor expression in the branching mouse lung. Dev Dyn 2012; 241:1432-53. [PMID: 22711520 PMCID: PMC3529173 DOI: 10.1002/dvdy.23823] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Mammalian lung development consists of a series of precisely choreographed events that drive the progression from simple lung buds to the elaborately branched organ that fulfills the vital function of gas exchange. Strict transcriptional control is essential for lung development. Among the large number of transcription factors encoded in the mouse genome, only a small portion of them are known to be expressed and function in the developing lung. Thus a systematic investigation of transcription factors expressed in the lung is warranted. RESULTS To enrich for genes that may be responsible for regional growth and patterning, we performed a screen using RNA in situ hybridization to identify genes that show restricted expression patterns in the embryonic lung. We focused on the pseudoglandular stage during which the lung undergoes branching morphogenesis, a cardinal event of lung development. Using a genome-scale probe set that represents over 90% of the transcription factors encoded in the mouse genome, we identified 62 transcription factor genes with localized expression in the epithelium, mesenchyme, or both. Many of these genes have not been previously implicated in lung development. CONCLUSIONS Our findings provide new starting points for the elucidation of the transcriptional circuitry that controls lung development.
Collapse
Affiliation(s)
- John C. Herriges
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Lan Yi
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Elizabeth A. Hines
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Julie F. Harvey
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Guoliang Xu
- Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China 200031
| | - Paul Gray
- Department of Anatomy and Neurobiology, Washington University, St. Louis, MO 63110
| | - Qiufu Ma
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
27
|
Pereira LA, Wong MS, Mei Lim S, Stanley EG, Elefanty AG. The Mix family of homeobox genes—Key regulators of mesendoderm formation during vertebrate development. Dev Biol 2012; 367:163-77. [DOI: 10.1016/j.ydbio.2012.04.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 04/24/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
|
28
|
Reid CD, Zhang Y, Sheets MD, Kessler DS. Transcriptional integration of Wnt and Nodal pathways in establishment of the Spemann organizer. Dev Biol 2012; 368:231-41. [PMID: 22627292 DOI: 10.1016/j.ydbio.2012.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/22/2012] [Accepted: 05/08/2012] [Indexed: 11/25/2022]
Abstract
Signaling inputs from multiple pathways are essential for the establishment of distinct cell and tissue types in the embryo. Therefore, multiple signals must be integrated to activate gene expression and confer cell fate, but little is known about how this occurs at the level of target gene promoters. During early embryogenesis, Wnt and Nodal signals are required for formation of the Spemann organizer, which is essential for germ layer patterning and axis formation. Signaling by both Wnt and Nodal pathways is required for the expression of multiple organizer genes, suggesting that integration of these signals is required for organizer formation. Here, we demonstrate transcriptional cooperation between the Wnt and Nodal pathways in the activation of the organizer genes Goosecoid (Gsc), Cerberus (Cer), and Chordin (Chd). Combined Wnt and Nodal signaling synergistically activates transcription of these organizer genes. Effectors of both pathways occupy the Gsc, Cer and Chd promoters and effector occupancy is enhanced with active Wnt and Nodal signaling. This suggests that, at organizer gene promoters, a stable transcriptional complex containing effectors of both pathways forms in response to combined Wnt and Nodal signaling. Consistent with this idea, the histone acetyltransferase p300 is recruited to organizer promoters in a Wnt and Nodal effector-dependent manner. Taken together, these results offer a mechanism for spatial and temporal restriction of organizer gene transcription by the integration of two major signaling pathways, thus establishing the Spemann organizer domain.
Collapse
Affiliation(s)
- Christine D Reid
- Department of Cell and Developmental Biology, University of Pennsylvania, School of Medicine, Room 1110 Biomedical Research Building 2/3, 421 Curie Boulevard, Philadelphia, PA 19104-6058, USA
| | | | | | | |
Collapse
|
29
|
Nodal-dependent mesendoderm specification requires the combinatorial activities of FoxH1 and Eomesodermin. PLoS Genet 2011; 7:e1002072. [PMID: 21637786 PMCID: PMC3102743 DOI: 10.1371/journal.pgen.1002072] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 03/23/2011] [Indexed: 12/24/2022] Open
Abstract
Vertebrate mesendoderm specification requires the Nodal signaling pathway and its transcriptional effector FoxH1. However, loss of FoxH1 in several species does not reliably cause the full range of loss-of-Nodal phenotypes, indicating that Nodal signals through additional transcription factors during early development. We investigated the FoxH1-dependent and -independent roles of Nodal signaling during mesendoderm patterning using a novel recessive zebrafish FoxH1 mutation called midway, which produces a C-terminally truncated FoxH1 protein lacking the Smad-interaction domain but retaining DNA–binding capability. Using a combination of gel shift assays, Nodal overexpression experiments, and genetic epistasis analyses, we demonstrate that midway more accurately represents a complete loss of FoxH1-dependent Nodal signaling than the existing zebrafish FoxH1 mutant schmalspur. Maternal-zygotic midway mutants lack notochords, in agreement with FoxH1 loss in other organisms, but retain near wild-type expression of markers of endoderm and various nonaxial mesoderm fates, including paraxial and intermediate mesoderm and blood precursors. We found that the activity of the T-box transcription factor Eomesodermin accounts for specification of these tissues in midway embryos. Inhibition of Eomesodermin in midway mutants severely reduces the specification of these tissues and effectively phenocopies the defects seen upon complete loss of Nodal signaling. Our results indicate that the specific combinations of transcription factors available for signal transduction play critical and separable roles in determining Nodal pathway output during mesendoderm patterning. Our findings also offer novel insights into the co-evolution of the Nodal signaling pathway, the notochord specification program, and the chordate branch of the deuterostome family of animals. Multiple signaling pathways function combinatorially to form and pattern the primary tissue layers of almost all organisms, by interacting with each other and by utilizing different pathway components to perform specific roles. Here we investigated the combinatorial aspects of the Nodal signaling pathway, which is essential for proper induction of mesoderm and endoderm in vertebrates. We identified a new mutation in the zebrafish FoxH1 gene, which encodes a Nodal pathway transcription factor, a protein that responds to Nodal signals to carry out the pathway's cellular functions by regulating target gene expression. Using this mutation, we determined that FoxH1 acts in a combinatorial fashion with two other transcription factors, called Mixer and Eomesodermin, to carry out all roles of the Nodal pathway during early development. Through genetic manipulation, we were able to identify the discrete functions regulated by different combinations of these three transcription factors. Our results indicate that the availability of specific Nodal-responsive transcription factors dictates the functions of the Nodal pathway in specific areas of the developing embryo. Our work also provides evidence that the FoxH1 family of transcription factors evolved concomitantly, and perhaps causally, with the chordate branch of animals, to which all vertebrates including humans belong.
Collapse
|
30
|
Siamois and Twin are redundant and essential in formation of the Spemann organizer. Dev Biol 2011; 352:367-81. [PMID: 21295564 DOI: 10.1016/j.ydbio.2011.01.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 01/11/2011] [Accepted: 01/25/2011] [Indexed: 02/03/2023]
Abstract
The Spemann organizer is an essential signaling center in Xenopus germ layer patterning and axis formation. Organizer formation occurs in dorsal blastomeres receiving both maternal Wnt and zygotic Nodal signals. In response to stabilized βcatenin, dorsal blastomeres express the closely related transcriptional activators, Siamois (Sia) and Twin (Twn), members of the paired homeobox family. Sia and Twn induce organizer formation and expression of organizer-specific genes, including Goosecoid (Gsc). In spite of the similarity of Sia and Twn sequence and expression pattern, it is unclear whether these factors function equivalently in promoter binding and subsequent transcriptional activation, or if Sia and Twn are required for all aspects of organizer function. Here we report that Sia and Twn activate Gsc transcription by directly binding to a conserved P3 site within the Wnt-responsive proximal element of the Gsc promoter. Sia and Twn form homodimers and heterodimers by direct homeodomain interaction and dimer forms are indistinguishable in both DNA-binding and activation functions. Sequential chromatin immunoprecipitation reveals that the endogenous Gsc promoter can be occupied by either Sia or Twn homodimers or Sia-Twn heterodimers. Knockdown of Sia and Twn together, but not individually, results in a failure of organizer gene expression and a disruption of axis formation, consistent with a redundant role for Sia and Twn in organizer formation. Furthermore, simultaneous knockdown of Sia and Twn blocks axis induction in response to ectopic Wnt signaling, demonstrating an essential role for Sia and Twn in mediating the transcriptional response to the maternal Wnt pathway. The results demonstrate the functional redundancy of Sia and Twn and their essential role in direct transcriptional responses necessary for Spemann organizer formation.
Collapse
|
31
|
Rankin SA, Kormish J, Kofron M, Jegga A, Zorn AM. A gene regulatory network controlling hhex transcription in the anterior endoderm of the organizer. Dev Biol 2011; 351:297-310. [PMID: 21215263 DOI: 10.1016/j.ydbio.2010.11.037] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 11/15/2010] [Accepted: 11/17/2010] [Indexed: 10/18/2022]
Abstract
The homeobox gene hhex is one of the earliest markers of the anterior endoderm, which gives rise to foregut organs such as the liver, ventral pancreas, thyroid, and lungs. The regulatory networks controlling hhex transcription are poorly understood. In an extensive cis-regulatory analysis of the Xenopus hhex promoter, we determined how the Nodal, Wnt, and BMP pathways and their downstream transcription factors regulate hhex expression in the gastrula organizer. We show that Nodal signaling, present throughout the endoderm, directly activates hhex transcription via FoxH1/Smad2 binding sites in the proximal -0.44 Kb promoter. This positive action of Nodal is suppressed in the ventral-posterior endoderm by Vent 1 and Vent2, homeodomain repressors that are induced by BMP signaling. Maternal Wnt/β-catenin on the dorsal side of the embryo cooperates with Nodal and indirectly activates hhex expression via the homeodomain activators Siamois and Twin. Siamois/Twin stimulate hhex transcription through two mechanisms: (1) they induce the expression of Otx2 and Lim1 and together Siamois, Twin, Otx2, and Lim1 appear to promote hhex transcription through homeobox sites in a Wnt-responsive element located between -0.65 to -0.55 Kb of the hhex promoter. (2) Siamois/Twin also induce the expression of the BMP-antagonists Chordin and Noggin, which are required to exclude Vents from the organizer allowing hhex transcription. This study reveals a complex network regulating anterior endoderm transcription in the early embryo.
Collapse
Affiliation(s)
- Scott A Rankin
- Division of Developmental Biology, Cincinnati Children's Research Foundation and Department of Pediatrics, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Yum J, Jeong HM, Kim S, Seo JW, Han Y, Lee KY, Yeo CY. PKA-mediated stabilization of FoxH1 negatively regulates ERalpha activity. Mol Cells 2009; 28:67-71. [PMID: 19711044 DOI: 10.1007/s10059-009-0099-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 05/27/2009] [Accepted: 05/28/2009] [Indexed: 01/29/2023] Open
Abstract
Estrogen receptor alpha (ERalpha) mediates the mitogenic effects of estrogen. ERalpha signaling regulates the normal growth and differentiation of mammary tissue, but uncontrolled ERalpha activation increases the risk to breast cancer. Estrogen binding induces ligand-dependent ERalpha activation, thereby facilitating ERalpha dimerization, promoter binding and coactivator recruitment. ERalpha can also be activated in a ligand-independent manner by many signaling pathways, including protein kinase A (PKA) signaling. However, in several ERalpha-positive breast cancer cells, PKA inhibits estrogen-dependent cell growth. FoxH1 represses the transcriptional activities of estrogen receptors and androgen receptors (AR). Interestingly, FoxH1 has been found to inhibit the PKA-induced and ligand-induced activation of AR. In the present study, we examined the effects of PKA activation on the ability of FoxH1 to represses ERalpha transcriptional activity. We found that PKA increases the protein stability of FoxH1, and that FoxH1 inhibits PKA-induced and estradiol-induced activation of an estrogen response element (ERE). Furthermore, in MCF7 cells, FoxH1 knockdown increased the PKA-induced and estradiol-induced activation of the ERE. These results suggest that PKA can negatively regulate ERalpha, at least in part, through FoxH1.
Collapse
Affiliation(s)
- Jinah Yum
- Department of Life Science and Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Kerr TC, Cuykendall TN, Luettjohann LC, Houston DW. Maternal Tgif1 regulates nodal gene expression in Xenopus. Dev Dyn 2008; 237:2862-73. [PMID: 18816846 DOI: 10.1002/dvdy.21707] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In Xenopus, the maternal transcription factor VegT is necessary and sufficient to initiate the expression of nodal-related genes, which are central to many aspects of early development. However, little is known about regulation of VegT activity. Using maternal loss-of-function experiments, we show that the maternal homeoprotein, Tgif1, antagonizes VegT and plays a central role in anteroposterior patterning by negatively regulating a subset of nodal-related genes. Depletion of Tgif1 causes the anteriorization of embryos and the up-regulation of nodal paralogues nr5 and nr6. Furthermore, Tgif1 inhibits activation of nr5 by VegT in a manner that requires a C-terminal Sin3 corepressor-interacting domain. Tgif1 has been implicated in the transcriptional corepression of transforming growth factor-beta (TGFbeta) and retinoid signaling. However, we show that Tgif1 does not inhibit these pathways in early development. These results identify an essential role for Tgif1 in the control of nodal expression and provide insight into Tgif1 function and mechanisms controlling VegT activity.
Collapse
Affiliation(s)
- Tyler C Kerr
- University of Iowa, Department of Biology, Iowa City, Iowa 52246-1324, USA
| | | | | | | |
Collapse
|
35
|
Baek S, Seeling JM. Identification of a novel conserved mixed-isoform B56 regulatory subunit and spatiotemporal regulation of protein phosphatase 2A during Xenopus laevis development. BMC DEVELOPMENTAL BIOLOGY 2007; 7:139. [PMID: 18093315 PMCID: PMC2257934 DOI: 10.1186/1471-213x-7-139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 12/19/2007] [Indexed: 01/26/2023]
Abstract
Background Wnt signaling is a key regulator of development and tumorigenesis. Protein phosphatase 2A (PP2A), which consists of a catalytic C, a structural A, and a regulatory B subunit, plays diverse roles in Wnt signaling through its B56 subunits. B56 is a multigene family encoding for proteins with a conserved core domain and divergent amino- and carboxy-termini. Ectopic B56α and B56γ reduce β-catenin abundance and B56α reduces Wnt-dependent transcription, suggesting that B56α and B56γ inhibit Wnt signaling. In contrast, B56ε is required for Wnt signaling. Knowledge of where and when B56 subunits are expressed during Xenopus development will aid in our understanding of their roles in Wnt signaling. Results We have undertaken expression analyses of B56α and B56γ in Xenopus laevis. We cloned Xenopus B56α; it is 88% identical to human B56α. Xenopus B56γ is 94% identical with human B56γ, however, a novel evolutionarily conserved mixed-isoform transcript was identified that contains a B56δ-like amino-terminal domain and a B56γ core domain. The B56δ-like variable domain exon is located upstream of the B56γ variable domain exon at the human B56γ locus, suggesting that the mixed-isoform transcript is due to alternative splicing. B56γ transcripts with different 3' ends were identified that lack or possess a 35 base pair sequence, resulting in either a transcript similar to human B56γ1, or an uncharacterized evolutionarily conserved sequence. Real time RT-PCR analyses revealed that B56α is expressed at moderate levels before the midblastula transition (MBT), at reduced levels during gastrulation and neurulation, and at high levels during organogenesis, while B56γ is expressed at low levels until organogenesis. B56α is enriched in the ventral hemisphere pre-MBT, while B56γ is ventrally enriched post-MBT. Aα, Aβ, Cα and Cβ are expressed in early Xenopus development, suggesting the presence of a functional heterotrimer. Conclusion Our data suggest that B56 functional diversity is achieved in part through the synthesis of a novel mixed-isoform B56δ/γ transcript. Our data also suggest that B56α functions pre-MBT, inhibiting Wnt signaling on the ventral side of the embryo, and again during organogenesis, while B56γ functions primarily post-MBT.
Collapse
Affiliation(s)
- Sungmin Baek
- Department of Biology, City University of New York, Queens College, 65-30 Kissena Blvd,, Flushing, NY 11367, USA.
| | | |
Collapse
|
36
|
Kennedy MW, Green KA, Ford RL, Andrews PG, Paterno GD, Gillespie LL, Kao KR. Regulation of the response to Nodal-mediated mesoderm induction by Xrel3. Dev Biol 2007; 311:383-95. [PMID: 17920056 DOI: 10.1016/j.ydbio.2007.08.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 08/20/2007] [Accepted: 08/22/2007] [Indexed: 11/15/2022]
Abstract
The Xenopus egg has a yolk-laden vegetal hemisphere juxtaposed to a darkly pigmented animal hemisphere. Mesoderm is derived from the marginal zone, located at the interface between the two hemispheres. The vegetal-most cells become endoderm and release TGF-beta-related factors, including the Xenopus Nodal related (Xnr) proteins, which diffuse to induce the marginal zone to form mesoderm. The remaining animal cells become ectoderm, but our understanding of the mechanisms that limit the response to induction is incomplete. In this study, we provide evidence to suggest that Xrel3, a member of the Rel/NF-kappaB family, plays a role in defining the boundary separating induced from uninduced cells by regulating Xnr-responsive gene transcription. Ectopic Xrel3 expressed in prospective mesoderm caused repression of mesoderm-specific genes resulting in loss-of-function phenotypes that were rescued by co-expression of Xnr2. Depletion of Xrel3 from embryos with antisense morpholinos increased Xnr-dependent transcription, broadened expression of the pan-mesoderm marker Xbra and sensitized animal cells to mesoderm induction by Xnr2. We propose that an additional component to the mechanism that differentiates the ectoderm from the mesoderm involves regulation of nodal-dependent gene transcription by Xrel3.
Collapse
Affiliation(s)
- Mark W Kennedy
- Terry Fox Cancer Research Labs, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
Howard L, Rex M, Clements D, Woodland HR. Regulation of the Xenopus Xsox17alpha(1) promoter by co-operating VegT and Sox17 sites. Dev Biol 2007; 310:402-15. [PMID: 17719026 PMCID: PMC2098691 DOI: 10.1016/j.ydbio.2007.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Revised: 07/03/2007] [Accepted: 07/23/2007] [Indexed: 11/23/2022]
Abstract
The gene encoding the Sox F-group transcription factor Xsox17α1 is specifically expressed throughout the entire region of the Xenopus blastula fated to become endoderm, and is important in controlling endodermal development. Xsox17α1 is a direct target of the maternal endodermal determinant VegT and of Sox17 itself. We have analysed the promoter of the Xenopus laevis Xsox17α1 gene by transgenesis, and have identified two important control elements which reside about 9 kb upstream at the start of transcription. These elements individually drive transgenic endodermal expression in the blastula and gastrula. One contains functional, cooperating VegT and Sox-binding consensus sites. The Sox sites in this region are occupied in vivo. The other responds to TGF-β signals like Activin or Nodals that act through Smad2/3. We propose that these two regions co-operate in regulating the early endodermal expression of the Xsox17α1 gene.
Collapse
|
38
|
Pei W, Noushmehr H, Costa J, Ouspenskaia MV, Elkahloun AG, Feldman B. An early requirement for maternal FoxH1 during zebrafish gastrulation. Dev Biol 2007; 310:10-22. [PMID: 17719025 PMCID: PMC2121100 DOI: 10.1016/j.ydbio.2007.07.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 06/26/2007] [Accepted: 07/11/2007] [Indexed: 12/17/2022]
Abstract
The Forkhead Box H1 (FoxH1) protein is a co-transcription factor recruited by phosphorylated Smad2 downstream of several TGFbetas, including Nodal-related proteins. We have reassessed the function of zebrafish FoxH1 using antisense morpholino oligonucleotides (MOs). MOs targeting translation of foxH1 disrupt embryonic epiboly movements during gastrulation and cause death on the first day of development. The FoxH1 morphant phenotype is much more severe than that of zebrafish carrying foxh1/schmalspur (sur) DNA-binding domain mutations, FoxH1 splice-blocking morphants or other Nodal pathway mutants, and it cannot be altered by concomitant perturbations in Nodal signaling. Apart from disrupting epiboly, FoxH1 MO treatment disrupts convergence and internalization movements. Late gastrula-stage FoxH1 morphants exhibit delayed mesoderm and endoderm marker gene expression and failed patterning of the central nervous system. Probing FoxH1 morphant RNA by microarray, we identified a cohort of five keratin genes--cyt1, cyt2, krt4, krt8 and krt18--that are normally transcribed in the embryo's enveloping layer (EVL) and which have significantly reduced expression in FoxH1-depleted embryos. Simultaneously disrupting these keratins with a mixture of MOs reproduces the FoxH1 morphant phenotype. Our studies thus point to an essential role for maternal FoxH1 and downstream keratins during gastrulation that is epistatic to Nodal signaling.
Collapse
Affiliation(s)
| | | | | | | | | | - Benjamin Feldman
- Corresponding Author: Benjamin Feldman, Ph.D., Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, 9000 Rockville Pike, Building 35, Room 1B 205, Bethesda, MD 20892, Tel: (301) 402-6690, Fax: (301) 496-7184, E-mail:
| |
Collapse
|
39
|
Santos EM, Workman VL, Paull GC, Filby AL, Van Look KJW, Kille P, Tyler CR. Molecular basis of sex and reproductive status in breeding zebrafish. Physiol Genomics 2007; 30:111-22. [PMID: 17374843 DOI: 10.1152/physiolgenomics.00284.2006] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The zebrafish ( Danio rerio) is used extensively as a model species for studies on vertebrate development and for assessing chemical effects on reproduction. Despite this, the molecular mechanisms controlling zebrafish reproduction are poorly understood. We analyzed the transcriptomic profiles of the gonads of individual zebrafish, using a 17k oligonucleotide microarray, to define the molecular basis of sex and reproductive status in sexually mature fish. The gonadal transcriptome differed substantially between sexes. Among the genes overexpressed in females, 11 biological processes were overrepresented including mitochondrion organization and biogenesis, and cell growth and/or maintenance. Among the genes overexpressed in males, six biological processes were overrepresented including protein biosynthesis and protein metabolism. Analysis of the expression of gene families known to be involved in reproduction identified a number of genes differentially expressed between ovaries and testes including a number of sox genes and genes belonging to the insulin-like growth factor and the activin-inhibin pathways. Real-time quantitative PCR confirmed the expression profiles for nine of the most differentially expressed genes and indicated that many transcripts are likely to be switched off in one of the sexes in the gonads of adult fish. Significant differences were seen between the gonad transcriptomes of individual reproductively active females reflecting their stage of maturation, whereas the testis transcriptomes were remarkably similar between individuals. In summary, we have identified molecular processes associated with (gonadal) sex specificity in breeding zebrafish and established a strong relationship between individual ovarian transcriptomes and reproductive status in females.
Collapse
Affiliation(s)
- E M Santos
- School of Biosciences, University of Exeter, Exeter, UK.
| | | | | | | | | | | | | |
Collapse
|
40
|
Yaklichkin S, Vekker A, Stayrook S, Lewis M, Kessler DS. Prevalence of the EH1 Groucho interaction motif in the metazoan Fox family of transcriptional regulators. BMC Genomics 2007; 8:201. [PMID: 17598915 PMCID: PMC1939712 DOI: 10.1186/1471-2164-8-201] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 06/28/2007] [Indexed: 11/23/2022] Open
Abstract
Background The Fox gene family comprises a large and functionally diverse group of forkhead-related transcriptional regulators, many of which are essential for metazoan embryogenesis and physiology. Defining conserved functional domains that mediate the transcriptional activity of Fox proteins will contribute to a comprehensive understanding of the biological function of Fox family genes. Results Systematic analysis of 458 protein sequences of the metazoan Fox family was performed to identify the presence of the engrailed homology-1 motif (eh1), a motif known to mediate physical interaction with transcriptional corepressors of the TLE/Groucho family. Greater than 50% of Fox proteins contain sequences with high similarity to the eh1 motif, including ten of the nineteen Fox subclasses (A, B, C, D, E, G, H, I, L, and Q) and Fox proteins of early divergent species such as marine sponge. The eh1 motif is not detected in Fox proteins of the F, J, K, M, N, O, P, R and S subclasses, or in yeast Fox proteins. The eh1-like motifs are positioned C-terminal to the winged helix DNA-binding domain in all subclasses except for FoxG proteins, which have an N-terminal motif. Two similar eh1-like motifs are found in the zebrafish FoxQ1 and in FoxG proteins of sea urchin and amphioxus. The identification of eh1-like motifs by manual sequence alignment was validated by statistical analyses of the Swiss protein database, confirming a high frequency of occurrence of eh1-like sequences in Fox family proteins. Structural predictions suggest that the majority of identified eh1-like motifs are short α-helices, and wheel modeling revealed an amphipathicity that supports this secondary structure prediction. Conclusion A search for eh1 Groucho interaction motifs in the Fox gene family has identified eh1-like sequences in greater than 50% of Fox proteins. The results predict a physical and functional interaction of TLE/Groucho corepressors with many members of the Fox family of transcriptional regulators. Given the functional importance of the eh1 motif in transcriptional regulation, our annotation of this motif in the Fox gene family will facilitate further study of the diverse transcriptional and regulatory roles of Fox family proteins.
Collapse
Affiliation(s)
- Sergey Yaklichkin
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, 1110 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Alexander Vekker
- Department of Economics, University of Pennsylvania, 328 McNeil Building, 3718 Locust Walk, Philadelphia, PA 19104, USA
| | - Steven Stayrook
- Department of Biochemistry and Biophysics, University of Pennsylvania School of Medicine, 813B Stellar-Chance Laboratories, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Mitchell Lewis
- Department of Biochemistry and Biophysics, University of Pennsylvania School of Medicine, 813B Stellar-Chance Laboratories, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Daniel S Kessler
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, 1110 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
41
|
Izzi L, Silvestri C, von Both I, Labbé E, Zakin L, Wrana JL, Attisano L. Foxh1 recruits Gsc to negatively regulate Mixl1 expression during early mouse development. EMBO J 2007; 26:3132-43. [PMID: 17568773 PMCID: PMC1914101 DOI: 10.1038/sj.emboj.7601753] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 05/21/2007] [Indexed: 01/10/2023] Open
Abstract
Mixl1 is a member of the Mix/Bix family of paired-like homeodomain proteins and is required for proper axial mesendoderm morphogenesis and endoderm formation during mouse development. Mix/Bix proteins are transcription factors that function in Nodal-like signaling pathways and are themselves regulated by Nodal. Here, we show that Foxh1 forms a DNA-binding complex with Smads to regulate transforming growth factor beta (TGFbeta)/Nodal-dependent Mixl1 gene expression. Whereas Foxh1 is commonly described as a transcriptional activator, we observed that Foxh1-null embryos exhibit expanded and enhanced Mixl1 expression during gastrulation, indicating that Foxh1 negatively regulates expression of Mixl1 during early mouse embryogenesis. We demonstrate that Foxh1 associates with the homeodomain-containing protein Goosecoid (Gsc), which in turn recruits histone deacetylases to repress Mixl1 gene expression. Ectopic expression of Gsc in embryoid bodies represses endogenous Mixl1 expression and this effect is dependent on Foxh1. As Gsc is itself induced in a Foxh1-dependent manner, we propose that Foxh1 initiates positive and negative transcriptional circuits to refine cell fate decisions during gastrulation.
Collapse
Affiliation(s)
- Luisa Izzi
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Terence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
| | - Cristoforo Silvestri
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Terence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
| | - Ingo von Both
- Program in Molecular Biology and Cancer, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Etienne Labbé
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Terence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
| | - Lise Zakin
- Howard Hughes Medical Institute and Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Jeffrey L Wrana
- Program in Molecular Biology and Cancer, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada
| | - Liliana Attisano
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Terence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Room 1008, 160 College Street, Toronto, Ontario, Canada M5S 3E1. Tel.: +1 416 946 3129; Fax: +1 416 978 8287; E-mail:
| |
Collapse
|
42
|
Yun CH, Choi SC, Park E, Kim SJ, Chung AS, Lee HK, Lee HJ, Han JK. Negative regulation of Activin/Nodal signaling by SRF during Xenopus gastrulation. Development 2007; 134:769-77. [PMID: 17259304 DOI: 10.1242/dev.02778] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Activin/Nodal signaling is essential for germ-layer formation and axial patterning during embryogenesis. Recent evidence has demonstrated that the intra- or extracellular inhibition of this signaling is crucial for ectoderm specification and correct positioning of mesoderm and endoderm. Here, we analyzed the function of Xenopus serum response factor (XSRF) in establishing germ layers during early development. XSRF transcripts are restricted to the animal pole ectoderm in Xenopus early embryos. Ectopic expression of XSRF RNA suppresses mesoderm induction, both in the marginal zone in vivo and caused by Activin/Nodal signals in animal caps. Dominant-negative mutant or antisense morpholino oligonucleotide-mediated inhibition of XSRF function expands the expression of mesendodermal genes toward the ectodermal territory and enhances the inducing activity of the Activin signal. SRF interacts with Smad2 and FAST-1, and inhibits the formation of the Smad2-FAST-1 complex induced by Activin. These results suggest that XSRF might act to ensure proper mesoderm induction in the appropriate region by inhibiting the mesoderm-inducing signals during early embryogenesis.
Collapse
Affiliation(s)
- Chang-Hyun Yun
- Natural Medicines Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-333, Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Choi J, Dong L, Ahn J, Dao D, Hammerschmidt M, Chen JN. FoxH1 negatively modulates flk1 gene expression and vascular formation in zebrafish. Dev Biol 2007; 304:735-44. [PMID: 17306248 PMCID: PMC1876740 DOI: 10.1016/j.ydbio.2007.01.023] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 12/22/2006] [Accepted: 01/16/2007] [Indexed: 10/23/2022]
Abstract
Flk1 is the major receptor for VEGF on endothelial cells. During embryogenesis, flk1 is required for both vasculogenesis and angiogenesis and abnormally elevated flk1 expression is often associated with pathological conditions in adults. While the biological function of flk1 has been studied extensively, very little is known about how the flk1 gene is regulated at the transcriptional level. Our transgenic study led to the identification of a flk1 endothelial enhancer positioned approximately 5 kb upstream of the flk1 translation initiation site. Binding sites for FoxH1, scl, ets and gata factors are found in the zebrafish flk1 endothelial enhancer, as well as in upstream sequences of mouse flk1 and human kdr genes, suggesting that the regulatory machinery for flk1/kdr is conserved from fish to mammals. The roles of scl, ets and gata factors in hemangioblasts have been well defined, but the significance of FoxH1 in vessel formation has not been explored previously. Here we show that FoxH1 binds to the flk1 endothelial enhancer in vitro and functions as a repressor for flk1 transcription in cultured cells. Consistent with these findings, the expression level of flk1 is elevated in embryos lacking both maternal and zygotic FoxH1. We further show that overexpression of FoxH1 has a negative effect on vascular formation that can be counteracted by the down-regulation of smad2 activity in zebrafish embryos. Taken together, our data provide the first evidence that flk1 is a direct target of FoxH1 and that FoxH1 is involved in vessel formation in zebrafish.
Collapse
Affiliation(s)
- Jayoung Choi
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
44
|
Evolution of the mechanisms and molecular control of endoderm formation. Mech Dev 2007; 124:253-78. [PMID: 17307341 DOI: 10.1016/j.mod.2007.01.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 12/24/2006] [Accepted: 01/03/2007] [Indexed: 01/13/2023]
Abstract
Endoderm differentiation and movements are of fundamental importance not only for subsequent morphogenesis of the digestive tract but also to enable normal patterning and differentiation of mesoderm- and ectoderm-derived organs. This review defines the tissues that have been called endoderm in different species, their cellular origin and their movements. We take a comparative approach to ask how signaling pathways leading to embryonic and extraembryonic endoderm differentiation have emerged in different organisms, how they became integrated and point to specific gaps in our knowledge that would be worth filling. Lastly, we address whether the gastrulation movements that lead to endoderm internalization are coupled with its differentiation.
Collapse
|
45
|
Zorn AM, Wells JM. Molecular Basis of Vertebrate Endoderm Development. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 259:49-111. [PMID: 17425939 DOI: 10.1016/s0074-7696(06)59002-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The embryonic endoderm gives rise to the epithelial lining of the digestive and respiratory systems and organs such as the thyroid, lungs, liver, gallbladder, and pancreas. Studies in Xenopus, zebrafish, and mice have revealed a conserved molecular pathway controlling vertebrate endoderm development. The TGFbeta/Nodal signaling pathway is at the top of this molecular hierarchy and controls the expression of a number of key transcription factors including Mix-like homeodomain proteins, Gata zinc finger factors, Sox HMG domain proteins, and Fox forkhead factors. Here we review the function of these molecules comparing and contrasting their roles in each model organism. Finally, we will describe how our understanding of the molecular pathway governing endoderm development in embryos is being used to differentiate embryonic stem cells in vitro along endodermal lineages, with the ultimate goal of making therapeutically useful tissue.
Collapse
Affiliation(s)
- Aaron M Zorn
- Division of Developmental Biology, Cincinnati Children's Hospital Research, Foundation and University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|
46
|
Abstract
Developmental biology teachers use the example of the frog embryo to introduce young scientists to the wonders of vertebrate development, and to pose the crucial question, 'How does a ball of cells become an exquisitely patterned embryo?'. Classical embryologists also recognized the power of the amphibian model and used extirpation and explant studies to explore early embryo polarity and to define signaling centers in blastula and gastrula stage embryos. This review revisits these early stages of Xenopus development and summarizes the recent explosion of information on the intrinsic and extrinsic factors that are responsible for the first phases of embryonic patterning.
Collapse
Affiliation(s)
- Janet Heasman
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, OH 45229-3039, USA.
| |
Collapse
|
47
|
Dingwell KS, Smith JC. Tes regulates neural crest migration and axial elongation in Xenopus. Dev Biol 2006; 293:252-67. [PMID: 16554046 DOI: 10.1016/j.ydbio.2006.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2006] [Accepted: 02/03/2006] [Indexed: 11/20/2022]
Abstract
Tes is a member of an emerging family of proteins sharing a set of protein motifs referred to as PET-LIM domains. PET-LIM proteins such as Prickle regulate cell behavior during gastrulation in Xenopus and zebrafish, and to ask whether Tes is also involved in controlling cell behavior, we isolated its Xenopus orthologue. Xtes is expressed as a maternal transcript that is maintained at low levels until neurula stages when expression is elevated in the head and axial structures. Depletion of Xtes leads to a foreshortened head and severe defects in axis elongation. The anterior defect is due in part to the inhibition of cranial neural crest migration while the defects in elongation may be due to perturbation of expression of XFGF8, Xdelta-1 and Xcad-3 and thereby to disruption of posterior somitogenesis. Finally, we note that simultaneous depletion of Xtes and Xenopus Prickle results in axial defects that are more severe than those resulting from depletion of Xtes alone, suggesting that the two proteins act together to control axial elongation.
Collapse
Affiliation(s)
- Kevin S Dingwell
- Wellcome Trust and Cancer Research UK Gurdon Institute of Cancer and Developmental Biology, and Department of Zoology, University of Cambridge, Cambridge CB2 1QR, UK
| | | |
Collapse
|
48
|
Abstract
How important is the contribution of mRNAs and proteins stored in the oocyte for determining the body plan of the Xenopus embryo? Here we review the current understanding of the roles of maternally supplied transcription factors, signaling molecules, and signaling regulators in establishing the ectoderm, mesoderm, and endoderm germ layers and the embryonic axes. Key essential asymmetries of VegT, Wnt11, and Ectodermin are described, as well as the complexity of maternal transcription factors that are involved in the initial expression of early zygotic genes.
Collapse
Affiliation(s)
- Janet Heasman
- Division of Developmental Biology ML7007, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
49
|
Cha YR, Takahashi S, Wright CVE. Cooperative non-cell and cell autonomous regulation of Nodal gene expression and signaling by Lefty/Antivin and Brachyury in Xenopus. Dev Biol 2006; 290:246-64. [PMID: 16405884 DOI: 10.1016/j.ydbio.2005.10.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 10/20/2005] [Accepted: 10/28/2005] [Indexed: 11/25/2022]
Abstract
Dynamic spatiotemporal expression of the nodal gene and its orthologs is involved in the dose-dependent induction and patterning of mesendoderm during early vertebrate embryogenesis. We report loss-of-function studies that define a high degree of synergistic negative regulation on the Xenopus nodal-related genes (Xnrs) by extracellular Xenopus antivin/lefty (Xatv/Xlefty)-mediated functional antagonism and Brachyury-mediated transcriptional suppression. A strong knockdown of Xlefty/Xatv function was achieved by mixing translation- and splicing-blocking morpholino oligonucleotides that target both the A and B alloalleles of Xatv. Secreted and cell-autonomous inhibitors of Xnr signaling were used to provide evidence that Xnr-mediated induction was inherently long-range in this situation in the large amphibian embryo, essentially being capable of spreading over the entire animal hemisphere. There was a greater expansion of the Organizer and mesendoderm tissues associated with dorsal specification than noted in previous Xatv knockdown experiments in Xenopus, with consequent exogastrulation and long-term maintenance of expanded axial tissues. Xatv deficiency caused a modest animal-ward expansion of the marginal zone expression territory of the Xnr1 and Xnr2 genes. In contrast, introducing inhibitory Xbra-En(R) fusion constructs into Xatv-deficient embryos caused a much larger increase in the level and spatial extent of Xnr expression. However, in both cases (Xatv/Xlefty-deficiency alone, or combined with Xbra interference), Xnr2 expression was constrained to the superficial cell layer, suggesting a fundamental tissue-specific competence in the ability to express Xnrs, an observation with direct implications regarding the induction of endodermal vs. mesodermal fates. Our experiments reveal a two-level suppressive mechanism for restricting the level, range, and duration of Xnr signaling via extracellular inhibition by Xatv/Xlefty coupled with potent indirect transcriptional repression by Xbra.
Collapse
Affiliation(s)
- Young Ryun Cha
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN 37232, USA
| | | | | |
Collapse
|
50
|
Takahashi S, Onuma Y, Yokota C, Westmoreland JJ, Asashima M, Wright CVE. Nodal-related geneXnr5 is amplified in theXenopus genome. Genesis 2006; 44:309-21. [PMID: 16791846 DOI: 10.1002/dvg.20217] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In Xenopus, six nodal-related genes (Xnrs) have been identified to date. We found numerous tandem duplications of Xnr5 in the Xenopus laevis and Xenopus tropicalis genomes that involve highly conserved copies of coding and regulatory regions. The duplicated versions of Xnr5 were expressed in both the superficial and deep layer of dorsal endoderm and in the deep layer of ventral endoderm, where the initial inducers of mesendoderm formation would be expected to be localized. Overexpression of secreted inhibitors of Xnrs led to a substantially enhanced transcription of the duplicated Xnr5 genes and Xnr6 in embryos. Therefore, Xnr5 and Xnr6 have a novel feedback loop to inhibit transcription of Xnr5 and Xnr6. These results suggest that the initialization of a strong Xnr5 and Xnr6 signal is enabled by the rapid transcription from multiple genes. The novel feedback loop may negatively regulate transcription of Xnr5s and Xnr6 to limit overproduction of these potent inducers, with the Xnr5/Xnr6 signal then activating positive (Xnrs) and negative (Xlefty) loops, which regulate the range of mesodermal tissues produced.
Collapse
Affiliation(s)
- Shuji Takahashi
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8240, USA
| | | | | | | | | | | |
Collapse
|