1
|
Cudak N, López-Delgado AC, Keil S, Knopf F. Fibroblast growth factor pathway component expression in the regenerating zebrafish fin. Gene Expr Patterns 2023; 48:119307. [PMID: 36841347 DOI: 10.1016/j.gep.2023.119307] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023]
Abstract
Adult zebrafish regenerate their appendages (fins) after amputation including the regeneration of bone structures (fin rays). Fibroblast growth factor (Fgf) signaling, which is involved in morphogenetic processes during development, has been shown to be essential for the process of fin regeneration. Moreover, mutations in Fgf pathway component genes lead to abnormal skeletal growth in teleosts and mammals, including humans, illustrating the importance of Fgf signaling in the growth control of tissues. Here, we revisited Fgf signaling pathway component expression by RNA in situ hybridization to test for the expression of about half of the ligands and all receptors of the pathway in the regenerating zebrafish fin. Expression patterns of fgf7, fgf10b, fgf12b, fgf17b and fgfr1b have not been reported in the literature before. We summarize and discuss known and novel localization of expression and find that all five Fgf receptors (fgfr1a, fgfr1b, fgfr2, fgfr3 and fgfr4) and most of the tested ligands are expressed in specific regions of the regenerate. Our work provides a basis to study domain specific functions of Fgf signaling in the regenerating teleost appendage.
Collapse
Affiliation(s)
- Nicole Cudak
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Keil
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany; Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
2
|
Liu Y, Kossack ME, McFaul ME, Christensen LN, Siebert S, Wyatt SR, Kamei CN, Horst S, Arroyo N, Drummond IA, Juliano CE, Draper BW. Single-cell transcriptome reveals insights into the development and function of the zebrafish ovary. eLife 2022; 11:e76014. [PMID: 35588359 PMCID: PMC9191896 DOI: 10.7554/elife.76014] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Zebrafish are an established research organism that has made many contributions to our understanding of vertebrate tissue and organ development, yet there are still significant gaps in our understanding of the genes that regulate gonad development, sex, and reproduction. Unlike the development of many organs, such as the brain and heart that form during the first few days of development, zebrafish gonads do not begin to form until the larval stage (≥5 days post-fertilization). Thus, forward genetic screens have identified very few genes required for gonad development. In addition, bulk RNA-sequencing studies that identify genes expressed in the gonads do not have the resolution necessary to define minor cell populations that may play significant roles in the development and function of these organs. To overcome these limitations, we have used single-cell RNA sequencing to determine the transcriptomes of cells isolated from juvenile zebrafish ovaries. This resulted in the profiles of 10,658 germ cells and 14,431 somatic cells. Our germ cell data represents all developmental stages from germline stem cells to early meiotic oocytes. Our somatic cell data represents all known somatic cell types, including follicle cells, theca cells, and ovarian stromal cells. Further analysis revealed an unexpected number of cell subpopulations within these broadly defined cell types. To further define their functional significance, we determined the location of these cell subpopulations within the ovary. Finally, we used gene knockout experiments to determine the roles of foxl2l and wnt9b for oocyte development and sex determination and/or differentiation, respectively. Our results reveal novel insights into zebrafish ovarian development and function, and the transcriptome profiles will provide a valuable resource for future studies.
Collapse
Affiliation(s)
- Yulong Liu
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Michelle E Kossack
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Matthew E McFaul
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Lana N Christensen
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Stefan Siebert
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Sydney R Wyatt
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Caramai N Kamei
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Samuel Horst
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Nayeli Arroyo
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Iain A Drummond
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Celina E Juliano
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| |
Collapse
|
3
|
Ghilardi A, Diana A, Bacchetta R, Santo N, Ascagni M, Prosperi L, Del Giacco L. Inner Ear and Muscle Developmental Defects in Smpx-Deficient Zebrafish Embryos. Int J Mol Sci 2021; 22:ijms22126497. [PMID: 34204426 PMCID: PMC8235540 DOI: 10.3390/ijms22126497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 12/20/2022] Open
Abstract
The last decade has witnessed the identification of several families affected by hereditary non-syndromic hearing loss (NSHL) caused by mutations in the SMPX gene and the loss of function has been suggested as the underlying mechanism. In the attempt to confirm this hypothesis we generated an Smpx-deficient zebrafish model, pointing out its crucial role in proper inner ear development. Indeed, a marked decrease in the number of kinocilia together with structural alterations of the stereocilia and the kinocilium itself in the hair cells of the inner ear were observed. We also report the impairment of the mechanotransduction by the hair cells, making SMPX a potential key player in the construction of the machinery necessary for sound detection. This wealth of evidence provides the first possible explanation for hearing loss in SMPX-mutated patients. Additionally, we observed a clear muscular phenotype consisting of the defective organization and functioning of muscle fibers, strongly suggesting a potential role for the protein in the development of muscle fibers. This piece of evidence highlights the need for more in-depth analyses in search for possible correlations between SMPX mutations and muscular disorders in humans, thus potentially turning this non-syndromic hearing loss-associated gene into the genetic cause of dysfunctions characterized by more than one symptom, making SMPX a novel syndromic gene.
Collapse
Affiliation(s)
- Anna Ghilardi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Alberto Diana
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Renato Bacchetta
- Department of Environmental Science and Policy, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Nadia Santo
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Miriam Ascagni
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Laura Prosperi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Luca Del Giacco
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
- Correspondence:
| |
Collapse
|
4
|
Domenichini A, Casari I, Simpson PV, Desai NM, Chen L, Dustin C, Edmands JS, van der Vliet A, Mohammadi M, Massi M, Falasca M. Rhenium N-heterocyclic carbene complexes block growth of aggressive cancers by inhibiting FGFR- and SRC-mediated signalling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:276. [PMID: 33287862 PMCID: PMC7720599 DOI: 10.1186/s13046-020-01777-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Platinum-based anticancer drugs have been at the frontline of cancer therapy for the last 40 years, and are used in more than half of all treatments for different cancer types. However, they are not universally effective, and patients often suffer severe side effects because of their lack of cellular selectivity. There is therefore a compelling need to investigate the anticancer activity of alternative metal complexes. Here we describe the potential anticancer activity of rhenium-based complexes with preclinical efficacy in different types of solid malignancies. METHODS Kinase profile assay of rhenium complexes. Toxicology studies using zebrafish. Analysis of the growth of pancreatic cancer cell line-derived xenografts generated in zebrafish and in mice upon exposure to rhenium compounds. RESULTS We describe rhenium complexes which block cancer proliferation in vitro by inhibiting the signalling cascade induced by FGFR and Src. Initially, we tested the toxicity of rhenium complexes in vivo using a zebrafish model and identified one compound that displays anticancer activity with low toxicity even in the high micromolar range. Notably, the rhenium complex has anticancer activity in very aggressive cancers such as pancreatic ductal adenocarcinoma and neuroblastoma. We demonstrate the potential efficacy of this complex via a significant reduction in cancer growth in mouse xenografts. CONCLUSIONS Our findings provide a basis for the development of rhenium-based chemotherapy agents with enhanced selectivity and limited side effects compared to standard platinum-based drugs.
Collapse
Affiliation(s)
- Alice Domenichini
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Ilaria Casari
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Peter V Simpson
- Curtin Institute of Functional Molecules and Interfaces, Department of Chemistry, Curtin University, Perth, WA, 6102, Australia
| | - Nima Maheshkumar Desai
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia
| | - Lingfeng Chen
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Christopher Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Jeanne S Edmands
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Moosa Mohammadi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Massimiliano Massi
- Curtin Institute of Functional Molecules and Interfaces, Department of Chemistry, Curtin University, Perth, WA, 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth, WA, 6102, Australia.
| |
Collapse
|
5
|
Liu Y, Wang H, Wen H, Shi Y, Zhang M, Qi X, Zhang K, Gong Q, Li J, He F, Hu Y, Li Y. First High-Density Linkage Map and QTL Fine Mapping for Growth-Related Traits of Spotted Sea bass (Lateolabrax maculatus). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:526-538. [PMID: 32424479 DOI: 10.1007/s10126-020-09973-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/28/2020] [Indexed: 06/11/2023]
Abstract
Possessing powerful adaptive capacity and a pleasant taste, spotted sea bass (Lateolabrax maculatus) has a broad natural distribution and is one of the most popular mariculture fish in China. However, the genetic improvement program for this fish is still in its infancy. Growth is the most economically important trait and is controlled by quantitative trait loci (QTL); thus, the identification of QTLs and genetic markers for growth-related traits is an essential step for the establishment of marker-assisted selection (MAS) breeding programs. In this study, we report the first high-density linkage map of spotted sea bass constructed by sequencing 333 F1 generation individuals in a full-sib family using 2b-RAD technology. A total of 6883 SNP markers were anchored onto 24 linkage groups, spanning 2189.96 cM with an average marker interval of 0.33 cM. Twenty-four growth-related QTLs, including 13 QTLs for body weight and 11 QTLs for body length, were successfully detected, with phenotypic variance explained (PVE) ranging from 5.1 to 8.6%. Thirty potential candidate growth-related genes surrounding the associated SNPs were involved in cell adhesion, cell proliferation, cytoskeleton reorganization, calcium channels, and neuromodulation. Notably, the fgfr4 gene was detected in the most significant QTL; this gene plays a pivotal role in myogenesis and bone growth. The results of this study may facilitate marker-assisted selection for breeding populations and establish the foundation for further genomic and genetic studies investigating spotted sea bass.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Haolong Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yue Shi
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361005, China
| | - Meizhao Zhang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qingli Gong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Jifang Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Feng He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yanbo Hu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
6
|
Expression pattern of the small muscle protein, X-linked (smpx) gene during zebrafish embryonic and larval developmental stages. Gene Expr Patterns 2020; 36:119110. [PMID: 32197943 DOI: 10.1016/j.gep.2020.119110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 11/22/2022]
Abstract
The small muscle protein, X-linked (SMPX) gene encodes a cytoskeleton-associated protein, highly expressed in both cardiac and skeletal muscles, as well as in fetal inner ears, with suggested roles as mechanotransductor. Recently, several mutations in the SMPX gene have been associated with X-chromosomal progressive deafness in human. However, very little information is known concerning the roles of SMPX, and no in-vivo models are currently available. Therefore, we characterized the zebrafish ortholog of SMPX to pave the way towards the establishment of a biotool for future functional studies. Despite the genome duplication occurred in the ancestry of teleosts, zebrafish retain only one copy of smpx which shares a high degree of similarity with the mammalian counterpart in terms of genomic organization, syntenic map, and encoded protein. RT-PCR, as well as whole-mount in-situ hybridization and immunofluorescence analyses, revealed that smpx is expressed in several embryonic areas starting from the 4-somite stage. Specifically, smpx mRNA marked the Kupffer's vesicle (KV), the somites, the myocardium, the hair cells of the anterior and the posterior macula of the inner ear, the pronephric ducts, and the muscles of the branchial arches, eyes and pectoral fins. According to our data, zebrafish smpx expression pattern closely resembles that observed in mouse and human, supporting the notion that zebrafish might represent a suitable in-vivo model to disclose the cellular and molecular mechanisms underlying the involvement of SMPX in development and disease.
Collapse
|
7
|
Liu SB, Lu LF, Lu XB, Li S, Zhang YA. Zebrafish FGFR3 is a negative regulator of RLR pathway to decrease IFN expression. FISH & SHELLFISH IMMUNOLOGY 2019; 92:224-229. [PMID: 31200068 DOI: 10.1016/j.fsi.2019.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/30/2019] [Accepted: 06/04/2019] [Indexed: 06/09/2023]
Abstract
Fibroblast growth factor receptor (FGFR) 3 is one of the four distinct membrane-spanning tyrosine kinases required for proper skeletal development. In fish, the role of FGFR3 is still unclear. In this article, we reveal that zebrafish FGFR3 is a negative regulator of interferon (IFN) production in the innate immune response by suppressing the activity of TANK-binding kinase 1 (TBK1) in the process of virus infection. qPCR experiments demonstrate that the transcriptional level of cellular FGFR3 was upregulated by infection with spring viremia of carp virus (SVCV), indicating that FGFR3 might be involved in the process of host cell response to viral infection. Then, overexpression of FGFR3 significantly impeded the IFN promoter activity induced by a stimulator. In addition, the capabilities of a retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) system to activate IFN promoter were decreased during the overexpression of FGFR3. Subsequently, FGFR3 decreased the phosphorylation of interferon regulatory factor 3 (IRF3) and mediator of IRF3 activation (MITA) by TBK1. These findings suggest that zebrafish FGFR3 is a negative regulator of IFN by attenuating the kinase activity of TBK1, leading to the suppression of IFN expression.
Collapse
Affiliation(s)
- Shu-Bo Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xiao-Bing Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
8
|
Wei W, Liu W, Serra S, Asa SL, Ezzat S. The breast cancer susceptibility FGFR2 provides an alternate mode of HER2 activation. Oncogene 2015:onc2014440. [PMID: 25639874 DOI: 10.1038/onc.2014.440] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 11/05/2014] [Accepted: 12/05/2014] [Indexed: 01/22/2023]
Abstract
Fibroblast growth factor receptor 2 (FGFR2) has been shown reproducibly in genome-wide association studies to be associated with increased breast cancer risk. Here we show that mouse mammary tumor virus-neu mice develop breast carcinomas with FGFR2 immunoreactivity that parallels HER2 expression. FGFR2 signaling promotes HER2 shedding through the metalloprotease ADAM10 leading to intracellular accumulation of the truncated p95HER2 protein. This is accompanied by enhanced HER2 signaling and diminished sensitivity to trastuzumab. Functionally, FGFR2 facilitates HER2-mediated cell proliferation, acinar growth in three-dimensional morphogenesis assays and promotes tumor progression in mouse xenografts. These data implicate FGFR2 in a novel mechanism of ErbB activation and demonstrate an important interaction between FGFR2 and HER2 in promoting breast cancer progression.Oncogene advance online publication, 2 February 2015; doi:10.1038/onc.2014.440.
Collapse
Affiliation(s)
- W Wei
- 1] Department of Medicine, University Health Network, Toronto, Ontario, Canada [2] Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - W Liu
- 1] Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada [2] Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - S Serra
- 1] Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada [2] Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - S L Asa
- 1] Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada [2] Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - S Ezzat
- 1] Department of Medicine, University Health Network, Toronto, Ontario, Canada [2] Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Lee Y, Manegold JE, Kim AD, Pouget C, Stachura DL, Clements WK, Traver D. FGF signalling specifies haematopoietic stem cells through its regulation of somitic Notch signalling. Nat Commun 2014; 5:5583. [PMID: 25428693 PMCID: PMC4271318 DOI: 10.1038/ncomms6583] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/16/2014] [Indexed: 01/07/2023] Open
Abstract
Hematopoietic stem cells (HSCs) derive from hemogenic endothelial cells of the primitive dorsal aorta (DA) during vertebrate embryogenesis. The molecular mechanisms governing this unique endothelial to hematopoietic transition remain unclear. Here, we demonstrate a novel requirement for fibroblast growth factor (FGF) signaling in HSC emergence. This requirement is non-cell-autonomous, and acts within the somite to bridge the Wnt and Notch signaling pathways. We previously demonstrated that Wnt16 regulates the somitic expression of two Notch ligands, deltaC (dlc) and deltaD (dld), whose combined function is required for HSC fate. How Wnt16 connects to Notch function has remained an open question. Our current studies demonstrate that FGF signaling, via FGF receptor 4 (Fgfr4), mediates a signal transduction pathway between Wnt16 and Dlc, but not Dld, to regulate HSC specification. Our findings demonstrate that FGF signaling acts as a key molecular relay within the developmental HSC niche to instruct HSC fate.
Collapse
Affiliation(s)
- Yoonsung Lee
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Jennifer E Manegold
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Albert D Kim
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Claire Pouget
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - David L Stachura
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Biological Sciences, California State University, Chico, California 95929, USA
| | - Wilson K Clements
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Hematology, St Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - David Traver
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
10
|
Mechanisms of pituitary tumorigenesis. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
11
|
Rohs P, Ebert AM, Zuba A, McFarlane S. Neuronal expression of fibroblast growth factor receptors in zebrafish. Gene Expr Patterns 2013; 13:354-61. [DOI: 10.1016/j.gep.2013.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 05/27/2013] [Accepted: 06/22/2013] [Indexed: 10/26/2022]
|
12
|
The breast cancer susceptibility gene product fibroblast growth factor receptor 2 serves as a scaffold for regulation of NF-κB signaling. Mol Cell Biol 2012; 32:4662-73. [PMID: 22988296 DOI: 10.1128/mcb.00935-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Fibroblast growth factor (FGF) receptor 2 (FGFR2) has been identified in genome-wide association studies to be associated with increased breast cancer risk; however, its mechanism of action remains unclear. Here we show that the two major FGFR2 alternatively spliced isoforms, FGFR2-IIIb and FGFR2-IIIc, interact with IκB kinase β and its downstream target, NF-κB. FGFR2 inhibits nuclear RelA/p65 NF-κB translocation and activity and reduces expression of dependent transcripts, including interleukin-6. These interactions result in diminished STAT3 phosphorylation and reduced breast cancer cell growth, motility, and invasiveness. FGFR2 also arrests the epithelial cell-to-mesenchymal cell transition (EMT), resulting in attenuated neoplastic growth in orthotopic xenografts of breast cancer cells. Our studies provide strong evidence for the protective effects of FGFR2 on tumor progression. We propose that FGFR2 serves as a scaffold for multiple components of the NF-κB signaling complex. Through these interactions, FGFR2 isoforms can respond to tissue-specific FGF signals to modulate epithelial cell-stromal cell communications in cancer progression.
Collapse
|
13
|
Huettl RE, Haehl T, Huber AB. Fasciculation and guidance of spinal motor axons in the absence of FGFR2 signaling. PLoS One 2012; 7:e41095. [PMID: 22815929 PMCID: PMC3398880 DOI: 10.1371/journal.pone.0041095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
During development, fibroblast growth factors (FGF) are essential for early patterning events along the anterior-posterior axis, conferring positional identity to spinal motor neurons by activation of different Hox codes. In the periphery, signaling through one of four fibroblast growth factor receptors supports the development of the skeleton, as well as induction and maintenance of extremities. In previous studies, FGF receptor 2 (FGFR2) was found to interact with axon bound molecules involved in axon fasciculation and extension, thus rendering this receptor an interesting candidate for the promotion of proper peripheral innervation. However, while the involvement of FGFR2 in limb bud induction has been extensively studied, its role during axon elongation and formation of distinct nervous projections has not been addressed so far. We show here that motor neurons in the spinal cord express FGFR2 and other family members during the establishment of motor connections to the forelimb and axial musculature. Employing a conditional genetic approach to selectively ablate FGFR2 from motor neurons we found that the patterning of motor columns and the expression patterns of other FGF receptors and Sema3A in the motor columns of mutant embryos are not altered. In the absence of FGFR2 signaling, pathfinding of motor axons is intact, and also fasciculation, distal advancement of motor nerves and gross morphology and positioning of axonal projections are not altered. Our findings therefore show that FGFR2 is not required cell-autonomously in motor neurons during the formation of initial motor projections towards limb and axial musculature.
Collapse
Affiliation(s)
- Rosa-Eva Huettl
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Teresa Haehl
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Andrea B. Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- * E-mail:
| |
Collapse
|
14
|
Guo M, Liu W, Serra S, Asa SL, Ezzat S. FGFR2 isoforms support epithelial-stromal interactions in thyroid cancer progression. Cancer Res 2012; 72:2017-27. [PMID: 22345151 DOI: 10.1158/0008-5472.can-11-3985] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alternate splicing yields two distinct isoforms of the fibroblast growth factor (FGF) receptor FGFR2-IIIb and FGFR2-IIIc varying their extracellular structure in human thyroid cancer, in which FGFR expression is commonly dysregulated. In this study, we characterized the function of these variants in modulating thyroid cancer behavior. Enforced expression of either FGFR2-IIIb or FGFR2-IIIc in thyroid epithelial cancer cells reduced expression of fibronectin, MAGE-A3 and MMP9, while increasing p21 and enhancing Rb dephosphorylation. Consistent with these tumor-suppressive properties, FGFR2-IIIb and FGFR2-IIIc each diminished invasive behavior in vitro and reduced tumor growth and metastasis in vivo. Notably, these effects contrasted with those produced by expression of these FGFR isoforms in fibroblasts, in which they both stimulated cell growth. Moreover, in xenograft tumors generated by coimplantation of epithelial and fibroblast cells expressing that same isoform, there was no significant effect on tumor progression. Conversely, FGFR2-IIIb expression in epithelial cells yielded higher FGF4/FGF7 expression that, in the presence of FGFR2-IIIc-expressing fibroblasts, enhanced tumor progression. Together, our findings highlight the importance of cellular context in assigning growth properties to growth factor receptor isoforms. More specifically, they show how alternative splicing of FGFR2 yields heteroisoforms critical to the growth-promoting actions of FGFs that exert distinct epithelial-stromal effects in thyroid cancer.
Collapse
Affiliation(s)
- Miao Guo
- The Ontario Cancer Institute, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
15
|
Kapsimali M, Kaushik AL, Gibon G, Dirian L, Ernest S, Rosa FM. Fgf signaling controls pharyngeal taste bud formation through miR-200 and Delta-Notch activity. Development 2011; 138:3473-84. [PMID: 21791527 DOI: 10.1242/dev.058669] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Taste buds, the taste sensory organs, are conserved in vertebrates and composed of distinct cell types, including taste receptor, basal/presynaptic and support cells. Here, we characterize zebrafish taste bud development and show that compromised Fgf signaling in the larva results in taste bud reduction and disorganization. We determine that Fgf activity is required within pharyngeal endoderm for formation of Calb2b(+) cells and reveal miR-200 and Delta-Notch signaling as key factors in this process. miR-200 knock down shows that miR-200 activity is required for taste bud formation and in particular for Calb2b(+) cell formation. Compromised delta activity in mib(-/-) dramatically reduces the number of Calb2b(+) cells and increases the number of 5HT(+) cells. Conversely, larvae with increased Notch activity and ascl1a(-/-) mutants are devoid of 5HT(+) cells, but have maintained and increased Calb2b(+) cells, respectively. These results show that Delta-Notch signaling is required for intact taste bud organ formation. Consistent with this, Notch activity restores Calb2b(+) cell formation in pharyngeal endoderm with compromised Fgf signaling, but fails to restore the formation of these cells after miR-200 knock down. Altogether, this study provides genetic evidence that supports a novel model where Fgf regulates Delta-Notch signaling, and subsequently miR-200 activity, in order to promote taste bud cell type differentiation.
Collapse
Affiliation(s)
- Marika Kapsimali
- Ecole Normale Supérieure, Institut de Biologie, 75005 Paris, France.
| | | | | | | | | | | |
Collapse
|
16
|
Simões FC, Peterkin T, Patient R. Fgf differentially controls cross-antagonism between cardiac and haemangioblast regulators. Development 2011; 138:3235-45. [PMID: 21750034 DOI: 10.1242/dev.059634] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fibroblast growth factor (Fgf) has been implicated in the control of heart size during development, although whether this is by controlling cell fate, survival or proliferation has not been clear. Here, we show that Fgf, without affecting survival or proliferation, acts during gastrulation to drive cardiac fate and restrict anterior haemangioblast fate in zebrafish embryos. The haemangioblast programme was thought to be activated before the cardiac programme and is repressive towards it, suggesting that activation by Fgf of the cardiac programme might be via suppression of the haemangioblast programme. However, we show that the cardiac regulator nkx2.5 can also repress the haemangioblast programme and, furthermore, that cardiac specification still requires Fgf signalling even when haemangioblast regulators are independently suppressed. We further show that nkx2.5 and the cloche candidate gene lycat are expressed during gastrulation and regulated by Fgf, and that nkx2.5 overexpression, together with loss of the lycat targets etsrp and scl can stably induce expansion of the heart. We conclude that Fgf controls cardiac and haemangioblast fates by the simultaneous regulation of haemangioblast and cardiac regulators. We propose that elevation of Fgf signalling in the anterior haemangioblast territory could have led to its recruitment into the heart field during evolution, increasing the size of the heart.
Collapse
Affiliation(s)
- Filipa Costa Simões
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford University, John Radcliffe Hospital, Headington OX3 9DS, UK
| | | | | |
Collapse
|
17
|
Ota S, Tonou-Fujimori N, Tonou-Fujimori N, Nakayama Y, Ito Y, Kawamura A, Yamasu K. FGF receptor gene expression and its regulation by FGF signaling during early zebrafish development. Genesis 2010; 48:707-16. [PMID: 20960516 DOI: 10.1002/dvg.20682] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 09/11/2010] [Accepted: 10/07/2010] [Indexed: 12/23/2022]
Abstract
The expression of all four fgfr genes was extensively examined throughout early embryogenesis of the zebrafish (Danio rerio). fgfr1 alone was expressed maternally throughout the blastoderm, and then zygotically in the anterior neural plate and presomitic mesoderm. fgfr4 expression was first detected in late blastulae and was gradually restricted to the brain. fgfr2 and fgfr3 expression were initiated in early and late gastrulae, respectively; fgfr2 was expressed in the anterior neural plate and somitic mesoderm, whereas fgfr3 was activated in the axial mesoderm and then in the midbrain and somitic mesoderm. During somitogenesis, each of these fgfr genes was expressed in a characteristic manner in the brain. Using an FGF signal inhibitor, dominant-negative FGF receptors and fgf8.1/fgf8a mutants, we found that fgfr expression is directly or indirectly regulated by FGF signaling during epiboly and at the end of somitogenesis, revealing the presence of an autoregulatory mechanism.
Collapse
Affiliation(s)
- Satoshi Ota
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Zhu X, Asa SL, Ezzat S. Genetic and epigenetic mechanisms down-regulate FGF receptor 2 to induce melanoma-associated antigen A in breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2333-43. [PMID: 20348248 DOI: 10.2353/ajpath.2010.091049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent genome-wide association studies have identified single nucleotide polymorphisms (SNPs) in the gene encoding fibroblast growth factor receptor 2 (FGFR2) as a risk factor for breast cancer. We examined the relationship between these intron 2 SNPs and gene expression in breast carcinomas. Primary breast tissue showed a common occurrence of these SNPs accompanied by FGFR2 expression in normal ductal epithelium. Unexpectedly, we found that FGFR2 mRNA and protein levels were reduced in microdissected cancer cells when compared with paired normal breast epithelium. FGFR2 down-regulation was associated with DNA methylation and loss-of-heterozygosity. Where FGFR2-IIIb was expressed in tumor cells, it was accompanied by up-regulation of the RNA-binding proteins ESRP1/2, consistent with splicing of this isoform. Reduction in FGFR2 was associated with re-expression of its putative target melanoma-associated antigen (MAGE-A) in primary carcinoma cells. Conversely, forced expression or activation of FGFR2-IIIb resulted in MAGE-A silencing. These data provide the first evidence for FGFR2 down-regulation in breast carcinomas harboring intron 2 SNPs. Our findings underscore the significance of epigenetic and somatic changes that can potentially modify the effects of germline polymorphisms in determining FGFR2 gene expression.
Collapse
Affiliation(s)
- Xuegong Zhu
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
19
|
Gates KP, Mentzer L, Karlstrom RO, Sirotkin HI. The transcriptional repressor REST/NRSF modulates hedgehog signaling. Dev Biol 2010; 340:293-305. [PMID: 20122919 DOI: 10.1016/j.ydbio.2010.01.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 01/13/2010] [Accepted: 01/22/2010] [Indexed: 10/19/2022]
Abstract
The spatial and temporal control of gene expression is key to generation of specific cellular fates during development. Studies of the transcriptional repressor REST/NRSF (RE1 Silencing Transcription Factor or Neural Restrictive Silencing Factor) have provided important insight into the role that epigenetic modifications play in differential gene expression. However, the precise function of REST during embryonic development is not well understood. We have discovered a novel interaction between zebrafish Rest and the Hedgehog (Hh) signaling pathway. We observed that Rest knockdown enhances or represses Hh signaling in a context-dependant manner. In wild-type embryos and embryos with elevated Hh signaling, Rest knockdown augments transcription of Hh target genes. Conversely, in contexts where Hh signaling is diminished, Rest knockdown has the opposite effect and Hh target gene expression is further attenuated. Epistatic analysis revealed that Rest interacts with the Hh pathway at a step downstream of Smo. Furthermore, we present evidence implicating the bifunctional, Hh signaling component Gli2a as key to the Rest modulation of the Hh response. The role of Rest as a regulator of Hh signaling has broad implications for many developmental contexts where REST and Hh signaling act.
Collapse
Affiliation(s)
- Keith P Gates
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | |
Collapse
|
20
|
Nakada C, Iida A, Tabata Y, Watanabe S. Forkhead transcription factor foxe1 regulates chondrogenesis in zebrafish. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2010; 312:827-40. [PMID: 19488987 DOI: 10.1002/jez.b.21298] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Forkhead transcription factor (Fox) e1 is a causative gene for Bamforth-Lazarus syndrome, which is characterized by hypothyroidism and cleft palate. Applying degenerate polymerase chain reaction using primers specific for the conserved forkhead domain, we identified zebrafish foxe1 (foxe1). Foxe1 is expressed in the thyroid, pharynx, and pharyngeal skeleton during development; strongly expressed in the gill and weakly expressed in the brain, eye, and heart in adult zebrafish. A loss of function of foxe1 by morpholino antisense oligo (MO) exhibited abnormal craniofacial development, shortening of Meckel's cartilage and the ceratohyals, and suppressed chondrycytic proliferation. However, at 27 hr post fertilization, the foxe1 MO-injected embryos showed normal dlx2, hoxa2, and hoxb2 expression, suggesting that the initial steps of pharyngeal skeletal development, including neural crest migration and specification of the pharyngeal arch occurred normally. In contrast, at 2 dpf, a severe reduction in the expression of sox9a, colIIaI, and runx2b, which play roles in chondrocytic proliferation and differentiation, was observed. Interestingly, fgfr2 was strongly upregulated in the branchial arches of the foxe1 MO-injected embryos. Unlike Foxe1-null mice, normal thyroid development in terms of morphology and thyroid-specific marker expression was observed in foxe1 MO-injected zebrafish embryos. Taken together, our results indicate that Foxe1 plays an important role in chondrogenesis during development of the pharyngeal skeleton in zebrafish, probably through regulation of fgfr2 expression. Furthermore, the roles reported for FOXE1 in mammalian thyroid development may have been acquired during evolution.
Collapse
Affiliation(s)
- Chisako Nakada
- Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
21
|
Zhu X, Asa SL, Ezzat S. Histone-acetylated control of fibroblast growth factor receptor 2 intron 2 polymorphisms and isoform splicing in breast cancer. Mol Endocrinol 2009; 23:1397-405. [PMID: 19497954 DOI: 10.1210/me.2009-0071] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent genome-wide association studies have identified fibroblast growth factor receptor (FGFR)2 as one of a few candidate genes linked with breast cancer susceptibility. In particular, the disease-predisposing allele of FGFR2 is inherited as a 7.5-kb region within intron 2 that harbors eight single nucleotide polymorphisms. The relationship between these single nucleotide polymorphisms and FGFR2 gene expression remains unclear. Here we show the common occurrence of polymorphisms within the intron 2 region in a panel of 10 breast cancer cell lines. High FGFR2-expressing cell lines such as MCF-7 cells displayed polymorphic sequences with constitutive histone acetylation at multiple intron 2 sequences harboring putative transcription binding sites. Knockdown of Runx2 or CCAAT enhancer binding protein beta in these cells resulted in diminished endogenous FGFR2 gene expression. In contrast FGFR2-negative MDA-231 cells were wild type and showed evidence of histone 3/4 deacetylation at the rs2981578, rs10736303, and rs7895676 disease-associated alleles that harbor binding sites for Runx2, estrogen receptor, and CCAAT enhancer binding protein beta, respectively. Histone deacetylation inhibition with trichostatin A resulted in enhanced acetylation at these intron 2 sites, an effect associated with robust FGFR2 reexpression. Isoform analysis proved reexpression of the FGFR2-IIIc variant the splicing of which was positively influenced by trichostatin A-mediated recruitment of the Fas-activated serine/threonine phosphoprotein survival protein. Our findings highlight the potential role of histone acetylation in modulating access to selected polymorphic sites within intron 2 as well as downstream splicing sites in generating variable FGFR2 levels and isoforms in breast cancer.
Collapse
Affiliation(s)
- Xuegong Zhu
- Department of Medicine, and the Endocrine Oncology Site Group, Princess Margaret Hospital, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | | | | |
Collapse
|
22
|
Chong SW, Korzh V, Jiang YJ. Myogenesis and molecules - insights from zebrafish Danio rerio. JOURNAL OF FISH BIOLOGY 2009; 74:1693-1755. [PMID: 20735668 DOI: 10.1111/j.1095-8649.2009.02174.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Myogenesis is a fundamental process governing the formation of muscle in multicellular organisms. Recent studies in zebrafish Danio rerio have described the molecular events occurring during embryonic morphogenesis and have thus greatly clarified this process, helping to distinguish between the events that give rise to fast v. slow muscle. Coupled with the well-known Hedgehog signalling cascade and a wide variety of cellular processes during early development, the continual research on D. rerio slow muscle precursors has provided novel insights into their cellular behaviours in this organism. Similarly, analyses on fast muscle precursors have provided knowledge of the behaviour of a sub-set of epitheloid cells residing in the anterior domain of somites. Additionally, the findings by various groups on the roles of several molecules in somitic myogenesis have been clarified in the past year. In this study, the authors briefly review the current trends in the field of research of D. rerio trunk myogenesis.
Collapse
Affiliation(s)
- S-W Chong
- Laboratory of Developmental Signalling and Patterning, Genes and Development Division, A STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore.
| | | | | |
Collapse
|
23
|
Regan JC, Concha ML, Roussigne M, Russell C, Wilson SW. An Fgf8-dependent bistable cell migratory event establishes CNS asymmetry. Neuron 2009; 61:27-34. [PMID: 19146810 PMCID: PMC2790412 DOI: 10.1016/j.neuron.2008.11.030] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 10/15/2008] [Accepted: 11/17/2008] [Indexed: 10/21/2022]
Abstract
Neuroanatomical and functional asymmetries are universal features of the vertebrate CNS, but how asymmetry is generated is unknown. Here we show that zebrafish fgf8 mutants do not elaborate forebrain asymmetries, demonstrated by the failure of the parapineal nucleus to migrate from its initial midline position to the left side of the brain. Local provision of Fgf8 restores the asymmetric migration of parapineal cells, usually to the left, irrespective of the location of the Fgf8 source. This laterality bias is due to left-sided Nodal signaling and when the bias in Nodal signaling is removed, parapineal cells migrate toward the source of Fgf8 protein. This study presents a mechanism for breaking neuroanatomical symmetry through Fgf8-dependent regulation of bistable left- or right-sided migration of the parapineal. The combined action of Fgf and Nodal signals ensures the establishment of neuroanatomical asymmetries with consistent laterality.
Collapse
Affiliation(s)
- Jennifer C. Regan
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Miguel L. Concha
- Laboratory of Experimental Ontogeny, Nucleus of Neural Morphogenesis, ICBM, University of Chile, Independencia 1027, 8380453 Santiago, Chile
| | - Myriam Roussigne
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Claire Russell
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Stephen W. Wilson
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
24
|
Ota S, Tonou-Fujimori N, Yamasu K. The roles of the FGF signal in zebrafish embryos analyzed using constitutive activation and dominant-negative suppression of different FGF receptors. Mech Dev 2009; 126:1-17. [DOI: 10.1016/j.mod.2008.10.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 10/17/2008] [Accepted: 10/23/2008] [Indexed: 12/22/2022]
|
25
|
Abstract
Epigenetically-mediated gene dysregulation is a common feature associated with human pituitary tumorigenesis. The mechanisms leading to these changes, however, remain largely unknown. In this review, we examine changes responsible for DNA and histone modifications as independent, butpotentially interrlated modes of communication effecting chromatin remodeling. The dynamic properties of the enzymes involved in these reactions is highlighted. We use the fibroblast growth factor receptor 2 (FGFR2) as a model through which the p53-regulating melanoma-associated antigen (MAGE) system is governing in pituitary cells. The pathogenetic and potential therapeutic implications are discussed.
Collapse
Affiliation(s)
- Shereen Ezzat
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Esterberg R, Fritz A. dlx3b/4b are required for the formation of the preplacodal region and otic placode through local modulation of BMP activity. Dev Biol 2008; 325:189-99. [PMID: 19007769 DOI: 10.1016/j.ydbio.2008.10.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 09/19/2008] [Accepted: 10/13/2008] [Indexed: 12/20/2022]
Abstract
The vertebrate inner ear arises from the otic placode, a transient thickening of ectodermal epithelium adjacent to neural crest domains in the presumptive head. During late gastrulation, cells fated to comprise the inner ear are part of a domain in cranial ectoderm that contain precursors of all sensory placodes, termed the preplacodal region (PPR). The combination of low levels of BMP activity coupled with high levels of FGF signaling are required to establish the PPR through induction of members of the six/eya/dach, iro, and dlx families of transcription factors. The zebrafish dlx3b/4b transcription factors are expressed at the neural plate border where they play partially redundant roles in the specification of the PPR, otic and olfactory placodes. We demonstrate that dlx3b/4b assist in establishing the PPR through the transcriptional regulation of the BMP antagonist cv2. Morpholino-mediated knockdown of Dlx3b/4b results in loss of cv2 expression in the PPR and a transient increase in Bmp4 activity that lasts throughout early somitogenesis. Through the cv2-mediated inhibition of BMP activity, dlx3b/4b create an environment where FGF activity is favorable for PPR and otic marker expression. Our results provide insight into the mechanisms of PPR specification as well as the role of dlx3b/4b function in PPR and otic placode induction.
Collapse
|
27
|
Zhu X, Asa SL, Ezzat S. Fibroblast growth factor 2 and estrogen control the balance of histone 3 modifications targeting MAGE-A3 in pituitary neoplasia. Clin Cancer Res 2008; 14:1984-96. [PMID: 18381936 DOI: 10.1158/1078-0432.ccr-07-2003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Four members of the fibroblast growth factor receptor (FGFR) family transduce signals of a diverse group of FGF ligands. The FGFR2-IIIb isoform is abundantly present in the normal pituitary gland with contrasting down-regulation in neoplastic pituitary cells. cDNA profiling identified the cancer-testis antigen melanoma-associated antigen A3 (MAGE-A3) as a putative target negatively regulated by FGFR2. EXPERIMENTAL DESIGN Comparisons were made between normal and neoplastic human and mouse pituitary cells. Gene expression was examined by reverse transcription-PCR, DNA methylation was determined by methylation-specific PCR and combined bisulfite restriction analysis, and histone modification marks were identified by chromatin immunoprecipitation. RESULTS Normal human pituitary tissue that expresses FGFR2-IIIb does not express MAGE-A3; in contrast, pituitary tumors that are FGFR2 negative show abundant MAGE-A3 mRNA expression. MAGE-A3 expression correlates with the presence and extent of DNA promoter methylation; more frequent and higher-degree methylation is present in the normal gland compared with pituitary tumors. Conversely, pituitary tumors are hypomethylated, particularly in females where MAGE-A3 expression is nearly thrice higher than in males. Estradiol treatment induces MAGE-A3 through enhanced histone 3 acetylation and diminished methylation. The effects of estradiol are directly opposed by FGF7/FGFR2-IIIb. Down-regulation of MAGE-A3 results in p53 transcriptional induction, also through reciprocal histone acetylation and methylation modifications. CONCLUSIONS These findings highlight MAGE-A3 as a target of FGFR2-IIIb and estrogen action and provide evidence for a common histone-modifying network in the control of the balance between opposing signals.
Collapse
Affiliation(s)
- Xuegong Zhu
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
28
|
Abstract
Fibroblast growth factors (FGFs) are secreted polypeptide growth factors. The FGF signaling system plays crucial roles in multiple developmental processes in vertebrates. The human FGF family comprises 22 members. Although 16 zebrafish fgfs have been reported, the zebrafish fgf family has not been well elucidated. We have identified 11 additional zebrafish fgfs by conducting a homology-based search in the zebrafish genome and cDNA databases. The zebrafish fgf family now comprises at least 27 members. By conducting phylogenetic and gene location analyses, we examined relationships of zebrafish fgf genes with human FGF genes. All the zebrafish orthologs of human FGFs except for FGF9 have been identified. Zebrafish fgf9 might have been lost from the genome during evolution. In addition, six paralogs of zebrafish fgf genes have been identified. The phylogenetic analysis suggests that the zebrafish fgf gene family can be divided into seven subfamilies. The zebrafish fgf subfamilies are essentially consistent with the human FGF subfamilies, although some include potential paralogs. As the zebrafish system has proved useful for studying gene functions and genetic diseases, the present findings will be useful for elucidation of roles of FGFs in zebrafish and humans.
Collapse
Affiliation(s)
- Nobuyuki Itoh
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan.
| | | |
Collapse
|
29
|
Bushell KM, Söllner C, Schuster-Boeckler B, Bateman A, Wright GJ. Large-scale screening for novel low-affinity extracellular protein interactions. Genome Res 2008; 18:622-30. [PMID: 18296487 DOI: 10.1101/gr.7187808] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Extracellular protein-protein interactions are essential for both intercellular communication and cohesion within multicellular organisms. Approximately a fifth of human genes encode membrane-tethered or secreted proteins, but they are largely absent from recent large-scale protein interaction datasets, making current interaction networks biased and incomplete. This discrepancy is due to the unsuitability of popular high-throughput methods to detect extracellular interactions because of the biochemical intractability of membrane proteins and their interactions. For example, cell surface proteins contain insoluble hydrophobic transmembrane regions, and their extracellular interactions are often highly transient, having half-lives of less than a second. To detect transient extracellular interactions on a large scale, we developed AVEXIS (avidity-based extracellular interaction screen), a high-throughput assay that overcomes these technical issues and can detect very transient interactions (half-lives <or= 0.1 sec) with a low false-positive rate. We used it to systematically screen for receptor-ligand pairs within the zebrafish immunoglobulin superfamily and identified novel ligands for both well-known and orphan receptors. Genes encoding receptor-ligand pairs were often clustered phylogenetically and expressed in the same or adjacent tissues, immediately implying their involvement in similar biological processes. Using AVEXIS, we have determined the first systematic low-affinity extracellular protein interaction network, supported by independent biological data. This technique will now allow large-scale extracellular protein interaction mapping in a broad range of experimental contexts.
Collapse
Affiliation(s)
- K Mark Bushell
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, United Kingdom
| | | | | | | | | |
Collapse
|
30
|
Zhu X, Lee K, Asa SL, Ezzat S. Epigenetic silencing through DNA and histone methylation of fibroblast growth factor receptor 2 in neoplastic pituitary cells. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1618-28. [PMID: 17456767 PMCID: PMC1854956 DOI: 10.2353/ajpath.2007.061111] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/06/2007] [Indexed: 01/07/2023]
Abstract
Four members of the fibroblast growth factor receptor (FGFR) family of tyrosine kinases transduce signals of a diverse group of more than 23 fibroblast growth factor (FGF) ligands. Each prototypic receptor is composed of three immunoglobulin-like extracellular domains, two of which are involved in ligand binding. Alternative RNA splicing of one of two exons results in two different forms of the second half of the third immunoglobulin-like domain, the IIIb or IIIc isoforms. The contribution of each receptor and their isoforms in tumorigenesis remains unknown. In the pituitary, FGFR2 is expressed primarily as the IIIb isoform in normal adenohypophysial cells. In contrast, FGFR2 is significantly down-regulated in mouse corticotroph AtT20 tumor cells where the 5' promoter is methylated. Treatment of AtT20 cells with 5'-azacytidine resulted in FGFR2 re-expression, mainly as the FGFR2-IIIb isoform. Chromatin immunoprecipitation revealed evidence of histone methylation, but not of deacetylation, in the silencing of FGFR2 in AtT20 cells. Exposure of these cells to the cognate FGFR2-IIIb ligand FGF-7 resulted in diminished Rb phosphorylation and accumulation of p21 and p27, indicating diminished cell cycle progression. Examination of primary human pituitary adenomas revealed FGFR2 down-regulation in 52% (11 of 21) of samples and FGFR2 promoter DNA methylation in 45% (10 of 22) of samples. These data highlight the contribution from DNA and histone methylation as epigenetic mechanisms responsible for FGFR2 silencing in pituitary neoplasia.
Collapse
Affiliation(s)
- Xuegong Zhu
- Department of Medicine, Mount Sinai Hospital and University of Toronto, Canada
| | | | | | | |
Collapse
|
31
|
Songhet P, Adzic D, Reibe S, Rohr KB. fgf1 is required for normal differentiation of erythrocytes in zebrafish primitive hematopoiesis. Dev Dyn 2007; 236:633-43. [PMID: 17219402 DOI: 10.1002/dvdy.21056] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Hematopoiesis in vertebrate development involves an embryonic, primitive wave and a later, definitive wave in which embryonic blood cells are replaced with adult blood cells. We here show that zebrafish fgf1 is involved in vivo in primitive hematopoiesis. Fibroblast growth factor-1 (FGF1) morpholino knockdown leads to abnormal accumulation of blood cells in the posterior intermediate cell mass at 32 hr postfertilization. Expression of the erythroid markers gata1 and ika, normally diminishing in differentiating erythrocytes at this stage, is maintained at abnormally high levels in primitive blood cells. The onset of erythrocyte differentiation as assessed by o-dianisidine staining is severely delayed. Most fgf1 morphants later recover to wild-type appearance, and primitive erythrocytes eventually differentiate. Zebrafish fgf1 is syntenic to human FGF1, which maps to a critically deleted region in human del(5q) syndrome posing an increased risk of leukemia to patients. As its knockdown in zebrafish changes expression of gata1, a gene involved in hematopoietic stem cell decisions, FGF1 should be considered to play a role in the pathogenesis of del(5q) syndrome.
Collapse
Affiliation(s)
- Pascal Songhet
- University of Cologne, Institute for Developmental Biology, Köln, Germany
| | | | | | | |
Collapse
|
32
|
Blak AA, Naserke T, Weisenhorn DMV, Prakash N, Partanen J, Wurst W. Expression of Fgf receptors 1, 2, and 3 in the developing mid- and hindbrain of the mouse. Dev Dyn 2005; 233:1023-30. [PMID: 15830353 DOI: 10.1002/dvdy.20386] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8) mediates the function of the midbrain-hindbrain organizer (MHO). FGF signals are transmitted by means of four known FGF receptors (FGFRs). Studies of Fgfr expression in early vertebrate development have shown that Fgfr1 is expressed along the entire neural tube, whereas Fgfr2 and Fgfr3 expression has been shown to spare the tissue adjacent to the MHO. The FGF8 signal from the MHO, therefore, was believed to be transmitted by FGFR1 exclusively. However, incongruent results from conditional mutants of Fgf8 and Fgfr1 in the midbrain-hindbrain (MHB) region contradict this hypothesis. Therefore, we reexamined the expression of the Fgfrs in this region. Fgfr1 is expressed all over the neural tube. Strikingly, Fgfr2 is expressed throughout the floor plate of the MHB region. In the basal plate, Fgfr2 directly abuts the Fgf8 expression domain at the MHO, anteriorly and posteriorly. Fgfr3 expression is in contact with the Fgf8 expression domain only in the rostroventral hindbrain. Based on these findings, we postulate a role for FGFR2 and FGFR3 in FGF signaling in the ventral midbrain and hindbrain.
Collapse
Affiliation(s)
- Alexandra A Blak
- GSF-National Research Center for Environment and Health, Institute of Developmental Genetics, Neuherberg, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Nechiporuk A, Linbo T, Raible DW. Endoderm-derived Fgf3 is necessary and sufficient for inducing neurogenesis in the epibranchial placodes in zebrafish. Development 2005; 132:3717-30. [PMID: 16077091 DOI: 10.1242/dev.01876] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In vertebrates, epibranchial placodes are transient ectodermal thickenings that contribute sensory neurons to the epibranchial ganglia. These ganglia innervate internal organs and transmit information on heart rate, blood pressure and visceral distension from the periphery to the central nervous system. Despite their importance, the molecular mechanisms that govern the induction and neurogenesis of the epibranchial placodes are only now being elucidated. In this study, we demonstrate that endoderm is required for neurogenesis of the zebrafish epibranchial placodes. Mosaic analyses confirm that endoderm is the source of the neurogenic signal. Using a morpholino knockdown approach, we find that fgf3 is required for the majority of placode cells to undergo neurogenesis. Tissue transplants demonstrate that fgf3 activity is specifically required in the endodermal pouches. Furthermore, ectopic fgf3 expression is sufficient for inducing phox2a-positive neurons in wild-type and endoderm-deficient embryos. Surprisingly, ectodermal foxi1 expression, a marker for the epibranchial placode precursors, is present in both endoderm-deficient embryos and fgf3 morphants, indicating that neither endoderm nor Fgf3 is required for initial placode induction. Based on these findings, we propose a model for epibranchial placode development in which Fgf3 is a major endodermal determinant required for epibranchial placode neurogenesis.
Collapse
Affiliation(s)
- Alexei Nechiporuk
- Department of Biological Structure, University of Washington, Seattle, WA 98195-7420, USA
| | | | | |
Collapse
|
34
|
Groves JA, Hammond CL, Hughes SM. Fgf8 drives myogenic progression of a novel lateral fast muscle fibre population in zebrafish. Development 2005; 132:4211-22. [PMID: 16120642 DOI: 10.1242/dev.01958] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Fibroblast growth factors (Fgfs) have long been implicated in regulating vertebrate skeletal muscle differentiation, but their precise role(s) in vivo remain unclear. Here, we show that Fgf8 signalling in the somite is required for myod expression and terminal differentiation of a subset of fast muscle cells in the zebrafish lateral somite. In the absence of Fgf8, lateral somite cells transiently express myf5 but fail to make muscle and remain in a dermomyotome-like state characterised by pax3 and meox expression. Slow muscle fibres form and commence normal migration in the absence of Fgf8, but fail to traverse the expanded undifferentiated lateral somite. The Fgf8-independent residual population of medial fast muscle fibres is not Hedgehog dependent. However, Fgf8-independent medial fast muscle precursors are lacking in floatinghead mutants,suggesting that they require another ventral midline-derived signal. We conclude that Fgf8 drives terminal differentiation of a specific population of lateral muscle precursor cells within the early somite.
Collapse
Affiliation(s)
- Julie A Groves
- MRC Centre for Developmental Neurobiology and Randall Division for Cell and Molecular Biophysics, New Hunt's House, King's College London, London SE1 1UL, UK
| | | | | |
Collapse
|
35
|
Oates AC, Mueller C, Ho RK. Cooperative function of deltaC and her7 in anterior segment formation. Dev Biol 2005; 280:133-49. [PMID: 15766754 PMCID: PMC2801430 DOI: 10.1016/j.ydbio.2005.01.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Revised: 01/12/2005] [Accepted: 01/12/2005] [Indexed: 11/25/2022]
Abstract
Segmentation of paraxial mesoderm in vertebrates is regulated by a genetic oscillator that manifests as a series of wavelike or cyclic gene expression domains in the embryo. In zebrafish, this oscillator involves members of the Delta/Notch intercellular signaling pathway, and its down-stream targets, the Her family of transcriptional repressors. Loss of function of any one of the genes of this system, such as her7, gives rise to segmentation defects in the posterior trunk and tail, concomitant with a disruption of cyclic expression domains, indicating that the oscillator is required for posterior segmentation. Control of segmentation in the anterior trunk, and its relationship to that of the posterior is, however, not yet well understood. A combined loss of the cyclic Her genes her1 and her7 disrupts segmentation of both anterior and posterior paraxial mesoderm, indicating that her genes function redundantly in anterior segmentation. To test whether this anterior redundancy is specific to the her gene family, or alternatively is a more global feature of the segmentation oscillator, we looked at anterior segmentation after morpholino knock down of the cyclic cell-surface Notch ligand deltaC (dlc), either alone or in combination with her7, or other Delta/Notch pathway genes. We find that dlc is required for coherence of wavelike expression domains of cyclic genes her1 and her7 and maintenance of their expression levels, as well as for cyclic transcription of dlc itself, confirming that dlc is a component of the segmentation oscillator. Dose dependent, posteriorly-restricted segmentation defects were seen in the dlc knock down, and in combination with the deltaD or notch1a mutants. However, combined reduction of function of dlc and her7 results in defective segmentation of both anterior and posterior paraxial mesoderm, and a failure of cyclic expression domains to initiate, similar to loss of both her genes. Thus, anterior segmentation requires the functions of both her and delta family members in a parallel manner, suggesting that the segmentation oscillator operates in paraxial mesoderm along the entire vertebrate axis.
Collapse
Affiliation(s)
- Andrew C Oates
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| | | | | |
Collapse
|
36
|
Cao Y, Zhao J, Sun Z, Zhao Z, Postlethwait J, Meng A. fgf17b, a novel member of Fgf family, helps patterning zebrafish embryos. Dev Biol 2004; 271:130-43. [PMID: 15196956 DOI: 10.1016/j.ydbio.2004.03.032] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Revised: 03/01/2004] [Accepted: 03/25/2004] [Indexed: 11/20/2022]
Abstract
Fibroblast growth factors (Fgfs) play important roles in the pattern formation of early vertebrate embryos. We have identified a zebrafish ortholog of human FGF17, named fgf17b. The first phase of fgf17b expression occurs in the blastodermal margin of late blastulae and in the embryonic shield of early gastrulae. The second phase starts after the onset of segmentation, mainly in the presomitic mesoderm and newly formed somites. Injection of fgf17b mRNA into one-cell embryos induces expression of the mesodermal marker no tail (ntl) and rescues ntl expression suppressed by overexpression of lefty1 (lft1). Overexpression of fgf17b dorsalizes zebrafish gastrulae by enhancing expression of chordin (chd), which is an antagonist of the ventralizing signals BMPs. In addition, overexpression of fgf17b posteriorizes the neuroectoderm. Simultaneous knockdown of fgf17b and fgf8 with antisense morpholinos results in reduction of chd and ntl. Knockdown of fgf17b can alleviate inhibitory effect of ectopic expression of fgf3 on otx1. These data together suggest that Fgf17b plays a role in early embryonic patterning. We also demonstrate that fgf17b and fgf8 have stronger mesoderm inducting activity than fgf3, whereas fgf17b and fgf3 have stronger activity in posteriorizing the neuroectoderm than fgf8. Like fgf8, activation of fgf17b expression depends on Nodal signaling.
Collapse
Affiliation(s)
- Ying Cao
- Department of Biological Sciences and Biotechnology, State Key Laboratory of Biomembrane & Membrane Biotechnology, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | |
Collapse
|
37
|
Fürthauer M, Van Celst J, Thisse C, Thisse B. Fgf signalling controls the dorsoventral patterning of the zebrafish embryo. Development 2004; 131:2853-64. [PMID: 15151985 DOI: 10.1242/dev.01156] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The establishment of dorsoventral (DV) patterning in vertebrate embryos depends on the morphogenic activity of a group of Tgfβ superfamily members, the bone morphogenetic proteins (Bmps) (which specify ventral cell fates), and on their interaction with their dorsally secreted cognate inhibitors chordin and noggin. In the zebrafish, genetic analysis has revealed that Bmp2b and Bmp7, as well as their antagonist chordin, are required for proper DV patterning. The expression of Bmp genes is initially activated in the whole blastula. Well before the beginning of gastrulation, Bmp gene expression progressively disappears from the dorsal side to become restricted to the ventral part of the embryo. We show that this early restriction of Bmp gene expression, which occurs independently of noggin and chordin, is an essential step in the establishment of DV patterning. The progressive ventral restriction of Bmp gene transcripts is coincident with the spreading of Fgf activity from the dorsal side of the embryo, suggesting that Fgf signalling is implicated in dorsal downregulation of Bmp gene expression. In accordance with this, activation of the Fgf/Ras/Mapk-signalling pathway inhibits ventral Bmp gene expression, thereby causing a dorsalisation of the embryo. Conversely,inhibition of Fgf signalling causes Bmp gene expression to expand dorsally,leading to an expansion of ventral cell fates. In accordance with an important role of Fgf signalling in the DV patterning of the zebrafish, we show that loss of Fgf8 function enhances the ventralisation of chordin-deficient embryos. Our results thereby demonstrate that pre-gastrula stage Fgf-signalling is essential to delimit the expression domain of the genes encoding the functional morphogen of the dorsoventral axis of the early zebrafish embryo.
Collapse
Affiliation(s)
- Maximilian Fürthauer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, CNRS/INSERM/ULP, 1 rue Laurent Fries, BP10142, CU de Strasbourg, 67404, Illkirch Cedex, France
| | | | | | | |
Collapse
|
38
|
Johnston IA, Manthri S, Smart A, Campbell P, Nickell D, Alderson R. Plasticity of muscle fibre number in seawater stages of Atlantic salmon in response to photoperiod manipulation. J Exp Biol 2003; 206:3425-35. [PMID: 12939373 DOI: 10.1242/jeb.00577] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atlantic salmon (Salmo salar L.) were fed to satiety and reared from approximately 60 g to 5000 g at ambient seawater temperatures. The effect of photoperiod manipulation on muscle growth was investigated from the start of the first sea winter. Continuous light treatment in winter/spring (1 November to 18 June) improved growth performance in fish, resulting in a 30% increase in mean body mass relative to the ambient photoperiod fish by 12 August, but had no effect on sexual maturation. Significant increases in body mass in the continuous light groups were observed after 126 days (P<0.01). The number of fast muscle fibres per trunk cross-section was determined in a subset of the fish and was 28.5% higher in the continuous light (799 x 10(3)) than the natural day length (644 x 10(3)) groups after only 40 days, corresponding to the period of decreasing natural day length. Subsequent rates of fibre recruitment were similar between treatments. At the end of the fibre recruitment phase of growth (combined June and August samples), the maximum number of fast muscle fibres was 23% higher in fish from the cages receiving continuous light (881 x 10(3)+/-32 x 10(3); N=19) than in the ambient photoperiod cages (717 x 10(3)+/-15 x 10(3); N=20) (P<0.001). Continuous light treatment was associated with a shift in the distribution of fibre diameters, reflecting the altered patterns of fibre recruitment. However, the mean rate of fibre hypertrophy showed no consistent difference between treatments. There was a linear relationship between the myonuclear content of isolated single fibres and fibre diameter. On average, there were 27% more myonuclei in 150 microm-diameter fibres in the continuous light (3118 myonuclei cm(-1)) than the ambient photoperiod (2448 myonuclei cm(-1)) fish. After 40 days, continuous light treatment resulted in a transient increase in the density of myogenic progenitor cells, identified using a c-met antibody, to a level 70% above that of fish exposed to natural light. It is suggested that short days inhibited the proliferation of myogenic progenitor cells and that this was overcome by transferring fish to continuous light, causing an increase in the number of times the myogenic precursor cells divided and/or a decrease in cell cycle time. The net increase in myogenic progenitor cells resulted in proportional increases in the number and myonuclear content of fibres. The subsequent hypertrophy of these additional fibres can explain the delayed increase in body mass observed with continuous light treatment.
Collapse
Affiliation(s)
- Ian A Johnston
- Gatty Marine Laboratory, School of Biology, University of St Andrews, St Andrews, Fife KY16 8LB, Scotland, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Walshe J, Mason I. Unique and combinatorial functions of Fgf3 and Fgf8 during zebrafish forebrain development. Development 2003; 130:4337-49. [PMID: 12900450 DOI: 10.1242/dev.00660] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Complex spatiotemporal expression patterns of fgf3 and fgf8 within the developing zebrafish forebrain suggest their involvement in its regionalisation and early development. These factors have unique and combinatorial roles during development of more posterior brain regions, and here we report similar findings for the developing forebrain. We show that Fgf8 and Fgf3 regulate different aspects of telencephalic development, and that Fgf3 alone is required for the expression of several telencephalic markers. Within the diencephalon, Fgf3 and Fgf8 act synergistically to pattern the ventral thalamus, and are implicated in the regulation of optic stalk formation, whereas loss of Fgf3 alone results in defects in ZLI development. Forebrain commissure formation was abnormal in the absence of either Fgf3 or Fgf8; however, most severe defects were observed in the absence of both. Defects were observed in patterning of both the midline territory, within which the commissures normally form, and neuronal populations, whose axons comprise the commissures. Analysis of embryos treated with an FGFR inhibitor suggests that continuous FGF signalling is required from gastrulation stages for normal forebrain patterning, and identifies additional requirements for FGFR activity.
Collapse
Affiliation(s)
- Jennifer Walshe
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | | |
Collapse
|
40
|
Affiliation(s)
- Stephen T Brown
- Gonda Department of Cell and Molecular Biology, House Ear Institute, 2100 West Third Street, Los Angeles, California 90057, USA
| | | | | |
Collapse
|
41
|
Tonou-Fujimori N, Takahashi M, Onodera H, Kikuta H, Koshida S, Takeda H, Yamasu K. Expression of the FGF receptor 2 gene (fgfr2) during embryogenesis in the zebrafish Danio rerio. Mech Dev 2002; 119 Suppl 1:S173-8. [PMID: 14516681 DOI: 10.1016/s0925-4773(03)00112-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We isolated a full-length cDNA clone for the zebrafish homologue of fibroblast growth factor receptor (FGFR) 2. The deduced protein sequence is typical of vertebrate FGFRs in that it has three Ig-like domains in the extracellular region. The expression of fgfr2 is initiated during epiboly in the paraxial mesoderm. During early somitogenesis, fgfr2 expression was noted in the anterior neural plate as well as in newly formed somites. Whereas fgfr2 expression in somites is transient, it increases in the central nervous system (CNS), i.e. in the ventral telencephalon, anterior diencephalon, midbrain, and respective rhombomeres of the hindbrain, from the mid-somitogenesis stage. The dorsal telencephalon and the region around the midbrain-hindbrain boundary are devoid of fgfr2 expression. Essentially the same expression pattern is observed until 48 h post-fertilization in the CNS, although rhombomeric expression in the hindbrain is progressively confined to narrower stripes. After somitogenesis, fgfr2 expression was also observed in the lens, hypochord, endoderm, and fin mesenchyme. We compared the expression of the four fgfr genes (fgfr1-4) in the CNS of zebrafish embryos and show that fgfr1 is the only fgfr gene that is expressed in the dorsal telencephalon and isthmic region from which expression of fgfr2-4 is absent.
Collapse
Affiliation(s)
- Noriko Tonou-Fujimori
- Department of Regulation Biology, Faculty of Science, Saitama University, 255 Shimo-Okubo, Saitama City, Saitama 338-8570, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Cai J, Wu Y, Mirua T, Pierce JL, Lucero MT, Albertine KH, Spangrude GJ, Rao MS. Properties of a fetal multipotent neural stem cell (NEP cell). Dev Biol 2002; 251:221-40. [PMID: 12435354 DOI: 10.1006/dbio.2002.0828] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multipotent neural stem cells (NSCs) present in the developing neural tube (E10.5, neuroepithelial cells; NEP) were examined for the expression of candidate stem cell markers, and the expression of these markers was compared with later appearing precursor cells (E14.5) that can be distinguished by the expression of embryonic neural cell adhesion molecule (E-NCAM) and A2B5. NEP cells possess gap junctions, express connexins, and appear to lack long cilia. Most candidate markers, including Nestin, Presenilin, Notch, and Numb, were expressed by both NEP cells as well as other cell populations. Fibroblast growth factor receptor 4 (FGFR4), Frizzled 9 (Fz9), and SRY box-containing gene 2 (Sox2) as assessed by immunocytochemistry and in situ hybridization are markers that appear to distinguish NSCs from other precursor cells. Neither Hoechst 33342 nor rhodamine-123 staining, telomerase (Tert) expression, telomerase activity, or breakpoint cluster region protein 1 (Bcrp1) transporter expression could be used to distinguish NEP stem cells from other dividing cells. NEP cells, however, lacked expression of several lineage markers that are expressed by later appearing cells. These included absence of expression of CD44, E-NCAM, A2B5, epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor-alpha (PDGFR alpha), suggesting that negative selection using cell surface epitopes could be used to isolate stem cell populations from mixed cultures of cells. Using mixed cultures of cells isolated from E14.5 stage embryos, we show that NEP cells can be enriched by depleting differentiating cells that express E-NCAM or A2B5 immunoreactivity. Overall, our results show that a spectrum of markers used in combination can reliably distinguish multipotent NSCs from other precursor cells as well as differentiated cells present in the CNS.
Collapse
Affiliation(s)
- Jingli Cai
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Maves L, Jackman W, Kimmel CB. FGF3 and FGF8 mediate a rhombomere 4 signaling activity in the zebrafish hindbrain. Development 2002; 129:3825-37. [PMID: 12135921 DOI: 10.1242/dev.129.16.3825] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The segmentation of the vertebrate hindbrain into rhombomeres is highly conserved, but how early hindbrain patterning is established is not well understood. We show that rhombomere 4 (r4) functions as an early-differentiating signaling center in the zebrafish hindbrain. Time-lapse analyses of zebrafish hindbrain development show that r4 forms first and hindbrain neuronal differentiation occurs first in r4. Two signaling molecules, FGF3 and FGF8, which are both expressed early in r4, are together required for the development of rhombomeres adjacent to r4, particularly r5 and r6. Transplantation of r4 cells can induce expression of r5/r6 markers, as can misexpression of either FGF3 or FGF8. Genetic mosaic analyses also support a role for FGF signaling acting from r4. Taken together, our findings demonstrate a crucial role for FGF-mediated inter-rhombomere signaling in promoting early hindbrain patterning and underscore the significance of organizing centers in patterning the vertebrate neural plate.
Collapse
Affiliation(s)
- Lisa Maves
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403-1254, USA.
| | | | | |
Collapse
|
44
|
Praul CA, Ford BC, Leach RM. Effect of fibroblast growth factors 1, 2, 4, 5, 6, 7, 8, 9, and 10 on avian chondrocyte proliferation. J Cell Biochem 2002; 84:359-66. [PMID: 11787065 DOI: 10.1002/jcb.1300] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It has been demonstrated that fibroblast growth factor receptors are key regulators of endochondral bone growth. However, it has not been determined what fibroblast growth factor ligand(s) (FGFs) are important in this process. This study sought to determine whether FGFs 1, 2, 4, 5, 6, 7, 8, 9, and 10 were capable of stimulating avian chondrocyte proliferation in vitro. We have found that FGFs 2, 4, and 9 strongly stimulate avian chondrocyte proliferation while FGFs 6 and 8 stimulate proliferation to a lesser extent. RT-PCR indicates that FGF-2 and FGF-4 are expressed in the postnatal avian epiphyseal growth plate (EGP) while FGF-8 and FGF-9 are not. Thus, FGF-2 and FGF-4 stimulate chondrocyte proliferation and are both present in the EGP. This suggests that FGF-2 and FGF-4 may be important ligands, in vivo, for the regulation of endochondral bone growth. These observations coupled with our observation that multiple avian FGF receptors (Cek1, Cek2, Cek3, and FREK) are expressed in proliferative chondrocytes highlights the complexity of FGF signaling pathways in postnatal endochondral bone growth.
Collapse
Affiliation(s)
- Craig A Praul
- Department of Poultry Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | |
Collapse
|
45
|
Ford-Perriss M, Abud H, Murphy M. Fibroblast growth factors in the developing central nervous system. Clin Exp Pharmacol Physiol 2001; 28:493-503. [PMID: 11422214 DOI: 10.1046/j.1440-1681.2001.03477.x] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
1. It is now clear that members of the fibroblast growth factor (FGF) family have multiple roles during the formation of the central nervous system (CNS). 2. There are at least 23 members of the FGF family and, of these, 10 are expressed in the developing CNS, along with four FGF receptors (FGFR-1-4). 3. The present review discusses the roles of these FGFs, with emphasis on FGF-2, FGF-8, FGF-15 and FGF-17. Fibroblast growth factors-2 and -15 are generally expressed throughout the developing CNS, whereas FGF-8 and FGF-17 are tightly localized to specific regions of the developing brain and are only expressed in the embryo during the early phases of proliferation and neurogenesis. 4. Expression studies on FGFRs in the chick and mouse indicate that FGFR-1 is most generally expressed, whereas FGFR-2 and FGFR-3 show highly localized but changing patterns of expression throughout CNS development. The FGFR-4 has been localized to the developing CNS in fish but not at a detailed level, as yet, in chick or mouse. 5. A picture is emerging from these studies that particular FGFs signal through specific receptors in a highly localized manner to regulate the development of different regions of the brain. 6. This picture has been demonstrated so far for the developing cortex (FGF-2-/- mice), the forebrain and midbrain (FGF-8 hypomorphs) and the cerebellum (FGF-17/FGF-8 mutant mice). In addition, generation of mutant animals deleted for FGFR-1 and FGFR-2b IIIb demonstrate their importance in FGF signalling. 7. However, there are significant gaps in our knowledge of the localization of members of the FGF family and their receptors. More detailed information on the spatio-temporal mapping of FGFs and FGFR isoforms is required in order to understand the molecular mechanisms through which FGFs signal.
Collapse
Affiliation(s)
- M Ford-Perriss
- Department of Anatomy and Cell Biology, The University of Melbourne, Victoria 3010, Australia.
| | | | | |
Collapse
|
46
|
Fürthauer M, Reifers F, Brand M, Thisse B, Thisse C. sprouty4 acts in vivo as a feedback-induced antagonist of FGF signaling in zebrafish. Development 2001; 128:2175-86. [PMID: 11493538 DOI: 10.1242/dev.128.12.2175] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In looking for novel factors involved in the regulation of the fibroblast growth factor (FGF) signaling pathway, we have isolated a zebrafish sprouty4 gene, based on its extensive similarities with the expression patterns of both fgf8 and fgf3. Through gain- and loss-of-function experiments, we demonstrate that Fgf8 and Fgf3 act in vivo to induce the expression of Spry4, which in turn can inhibit activity of these growth factors. When overexpressed at low doses, Spry4 induces loss of cerebellum and reduction in size of the otic vesicle, thereby mimicking the fgf8/acerebellar mutant phenotype. Injections of high doses of Spry4 cause ventralization of the embryo, an opposite phenotype to the dorsalisation induced by overexpression of Fgf8 or Fgf3. Conversely we have shown that inhibition of Spry4 function through injection of antisense morpholino oligonucleotide leads to a weak dorsalization of the embryo, the phenotype expected for an upregulation of Fgf8 or Fgf3 signaling pathway. Finally, we show that Spry4 interferes with FGF signaling downstream of the FGF receptor 1 (FGFR1). In addition, our analysis reveals that signaling through FGFR1/Ras/mitogen-activated protein kinase pathway is involved, not in mesoderm induction, but in the control of the dorsoventral patterning via the regulation of bone morphogenetic protein (BMP) expression.
Collapse
Affiliation(s)
- M Fürthauer
- Institut de Génétique et Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, BP 163, 67404 Illkirch cedex, CU de Strasbourg, France
| | | | | | | | | |
Collapse
|
47
|
Christiansen JH, Coles EG, Robinson V, Pasini A, Wilkinson DG. Screening from a subtracted embryonic chick hindbrain cDNA library: identification of genes expressed during hindbrain, midbrain and cranial neural crest development. Mech Dev 2001; 102:119-33. [PMID: 11287186 DOI: 10.1016/s0925-4773(01)00294-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The vertebrate hindbrain is segmented into a series of transient structures called rhombomeres. Despite knowing several factors that are responsible for the segmentation and maintenance of the rhombomeres, there are still large gaps in understanding the genetic pathways that govern their development. To find previously unknown genes that are expressed within the embryonic hindbrain, a subtracted chick hindbrain cDNA library has been made and 445 randomly picked clones from this library have been analysed using whole mount in situ hybridisation. Thirty-six of these clones (8%) display restricted expression patterns within the hindbrain, midbrain or cranial neural crest and of these, twenty-two are novel and eleven encode peptides that correspond to or are highly related to proteins with previously uncharacterised roles during early neural development. The large proportion of genes with restricted expression patterns and previously unknown functions in the embryonic brain identified during this screen provides insights into the different types of molecules that have spatially regulated expression patterns in cranial neural tissue.
Collapse
Affiliation(s)
- J H Christiansen
- Division of Developmental Neurobiology, National Institute for Medical Research, The Ridgeway, Mill Hill, NW7 1AA, London, UK
| | | | | | | | | |
Collapse
|
48
|
Abstract
Fyn protein tyrosine kinase is present in the unfertilized and fertilized egg, becomes activated within minutes following fertilization, and has been localized to the cortical cytoplasm and spindle apparatus of the zygote. In order to establish the expression pattern of Fyn in the early embryo, we examined the distribution pattern of Fyn by immunofluorescence microscopy. Fyn protein is distributed evenly among cells of the cleavage stage zebrafish embryo and is concentrated in the cortical region of each cell. During blastula and gastrula stages, Fyn was expressed in all cells, however a subpopulation of cells exhibited strong nuclear staining for Fyn. Nuclear Fyn staining was not observed after the gastrula period of development, nor in the adult zebrafish. Immunoprecipitation of Fyn from isolated mid-blastula nuclei confirmed Fyn was present in the nucleus. This is, to our knowledge, the first demonstration of Fyn kinase, which lacks a nuclear localization signal, present in the nucleus. The transient compartmentalization of Fyn in the nucleus could be important in nuclear signaling.
Collapse
Affiliation(s)
- B J Rongish
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | |
Collapse
|
49
|
Poss KD, Shen J, Nechiporuk A, McMahon G, Thisse B, Thisse C, Keating MT. Roles for Fgf signaling during zebrafish fin regeneration. Dev Biol 2000; 222:347-58. [PMID: 10837124 DOI: 10.1006/dbio.2000.9722] [Citation(s) in RCA: 249] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Following amputation of a urodele limb or teleost fin, the formation of a blastema is a crucial step in facilitating subsequent regeneration. Using the zebrafish caudal fin regeneration model, we have examined the hypothesis that fibroblast growth factors (Fgfs) initiate blastema formation from fin mesenchyme. We find that fibroblast growth factor receptor 1 (fgfr1) is expressed in mesenchymal cells underlying the wound epidermis during blastema formation and in distal blastemal tissue during regenerative outgrowth. fgfr1 transcripts colocalize with those of msxb and msxc, putative markers for undifferentiated, proliferating cells. A zebrafish Fgf member, designated wfgf, is expressed in the regeneration epidermis during outgrowth. Furthermore, we show that a specific inhibitor of Fgfr1 applied immediately following fin amputation blocks blastema formation, without obvious effects on wound healing. This inhibitor blocks the proliferation of blastemal cells and the onset of msx gene transcription. Inhibition of Fgf signaling during ongoing fin regeneration prevents further outgrowth while downregulating the established expression of blastemal msx genes and epidermal sonic hedgehog. Our findings indicate that zebrafish fin blastema formation and regenerative outgrowth require Fgf signaling.
Collapse
Affiliation(s)
- K D Poss
- Howard Hughes Medical Institute, Eccles Institute of Human Genetics, University of Utah Health Sciences Center, 15N 2030E, Salt Lake City, Utah, 84112, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Reifers F, Walsh EC, Léger S, Stainier DY, Brand M. Induction and differentiation of the zebrafish heart requires fibroblast growth factor 8 (fgf8/acerebellar). Development 2000; 127:225-35. [PMID: 10603341 DOI: 10.1242/dev.127.2.225] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vertebrate heart development is initiated from bilateral lateral plate mesoderm that expresses the Nkx2.5 and GATA4 transcription factors, but the extracellular signals specifying heart precursor gene expression are not known. We describe here that the secreted signaling factor Fgf8 is expressed in and required for development of the zebrafish heart precursors, particularly during initiation of cardiac gene expression. fgf8 is mutated in acerebellar (ace) mutants, and homozygous mutant embryos do not establish normal circulation, although vessel formation is only mildly affected. In contrast, heart development, in particular of the ventricle, is severely abnormal in acerebellar mutants. Several findings argue that Fgf8 has a direct function in development of cardiac precursor cells: fgf8 is expressed in cardiac precursors and later in the heart ventricle. Fgf8 is required for the earliest stages of nkx2.5 and gata4, but not gata6, expression in cardiac precursors. Cardiac gene expression is restored in acerebellar mutant embryos by injecting fgf8 RNA, or by implanting a Fgf8-coated bead into the heart primordium. Pharmacological inhibition of Fgf signalling during formation of the heart primordium phenocopies the acerebellar heart phenotype, confirming that Fgf signaling is required independently of earlier functions during gastrulation. These findings show that fgf8/acerebellar is required for induction and patterning of myocardial precursors.
Collapse
Affiliation(s)
- F Reifers
- Department of Neurobiology, University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|