1
|
Maekawa M, Saito S, Isobe D, Takemoto K, Miura Y, Dobashi Y, Yamasu K. The Oct4-related PouV gene, pou5f3, mediates isthmus development in zebrafish by directly and dynamically regulating pax2a. Cells Dev 2024; 179:203933. [PMID: 38908828 DOI: 10.1016/j.cdev.2024.203933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Using a transgenic zebrafish line harboring a heat-inducible dominant-interference pou5f3 gene (en-pou5f3), we reported that this PouV gene is involved in isthmus development at the midbrain-hindbrain boundary (MHB), which patterns the midbrain and cerebellum. Importantly, the functions of pou5f3 reportedly differ before and after the end of gastrulation. In the present study, we examined in detail the effects of en-pou5f3 induction on isthmus development during embryogenesis. When en-pou5f3 was induced around the end of gastrulation (bud stage), the isthmus was abrogated or deformed by the end of somitogenesis (24 hours post-fertilization). At this stage, the expression of MHB markers -- such as pax2a, fgf8a, wnt1, and gbx2 -- was absent in embryos lacking the isthmus structure, whereas it was present, although severely distorted, in embryos with a deformed isthmus. We further found that, after en-pou5f3 induction at late gastrulation, pax2a, fgf8a, and wnt1 were immediately and irreversibly downregulated, whereas the expression of en2a and gbx2 was reduced only weakly and slowly. Induction of en-pou5f3 at early somite stages also immediately downregulated MHB genes, particularly pax2a, but their expression was restored later. Overall, the data suggested that pou5f3 directly upregulates at least pax2a and possibly fgf8a and wnt1, which function in parallel in establishing the MHB, and that the role of pou5f3 dynamically changes around the end of gastrulation. We next examined the transcriptional regulation of pax2a using both in vitro and in vivo reporter analyses; the results showed that two upstream 1.0-kb regions with sequences conserved among vertebrates specifically drove transcription at the MHB. These reporter analyses confirmed that development of the isthmic organizer is regulated by PouV through direct regulation of pax2/pax2a in vertebrate embryos.
Collapse
Affiliation(s)
- Masato Maekawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Shinji Saito
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Institute for Vaccine Research and Development, Hokkaido University, N21, W11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Daiki Isobe
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kazumasa Takemoto
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Road, U3156, Storrs, CT 06269, USA
| | - Yuhei Miura
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Yurie Dobashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| |
Collapse
|
2
|
Shi X, Yang Y, Ma X, Zhou Y, Guo Z, Wang C, Liu J. Probabilistic cell/domain-type assignment of spatial transcriptomics data with SpatialAnno. Nucleic Acids Res 2023; 51:e115. [PMID: 37941153 PMCID: PMC10711557 DOI: 10.1093/nar/gkad1023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 10/03/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023] Open
Abstract
In the analysis of both single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) data, classifying cells/spots into cell/domain types is an essential analytic step for many secondary analyses. Most of the existing annotation methods have been developed for scRNA-seq datasets without any consideration of spatial information. Here, we present SpatialAnno, an efficient and accurate annotation method for spatial transcriptomics datasets, with the capability to effectively leverage a large number of non-marker genes as well as 'qualitative' information about marker genes without using a reference dataset. Uniquely, SpatialAnno estimates low-dimensional embeddings for a large number of non-marker genes via a factor model while promoting spatial smoothness among neighboring spots via a Potts model. Using both simulated and four real spatial transcriptomics datasets from the 10x Visium, ST, Slide-seqV1/2, and seqFISH platforms, we showcase the method's improved spatial annotation accuracy, including its robustness to the inclusion of marker genes for irrelevant cell/domain types and to various degrees of marker gene misspecification. SpatialAnno is computationally scalable and applicable to SRT datasets from different platforms. Furthermore, the estimated embeddings for cellular biological effects facilitate many downstream analyses.
Collapse
Affiliation(s)
- Xingjie Shi
- KLATASDS-MOE, Academy of Statistics and Interdisciplinary Sciences, School of Statistics, East China Normal University, Shanghai 200062, China
| | - Yi Yang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210018, China
| | - Xiaohui Ma
- College of Life Sciences, Nanjing University, Nanjing 210033, China
| | - Yong Zhou
- KLATASDS-MOE, Academy of Statistics and Interdisciplinary Sciences, School of Statistics, East China Normal University, Shanghai 200062, China
| | - Zhenxing Guo
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Jin Liu
- School of Data Science, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China
| |
Collapse
|
3
|
Offen N, Filatova A, Nuber UA. Enrichment of FGF8-expressing cells from neurally induced human pluripotent stem cell cultures. Stem Cell Reports 2023; 18:2240-2253. [PMID: 37922914 PMCID: PMC10679777 DOI: 10.1016/j.stemcr.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
In early vertebrate development, organizer regions-groups of cells that signal to and thereby influence neighboring cells by secreted morphogens-play pivotal roles in the establishment and maintenance of cell identities within defined tissue territories. The midbrain-hindbrain organizer drives regionalization of neural tissue into midbrain and hindbrain territories with fibroblast growth factor 8 (FGF8) acting as a key morphogen. This organizer has been extensively studied in chicken, mouse, and zebrafish. Here, we demonstrate the enrichment of FGF8-expressing cells from human pluripotent stem cells (hPSCs), cultured as attached embryoid bodies using antibodies that recognize "Similar Expression to Fgf" (SEF) and Frizzled proteins. The arrangement of cells in embryoid body subsets of these cultures and the gene expression profile of the FGF8-expressing population show certain similarities to the midbrain-hindbrain organizer in animal models. In the embryonic chick brain, the enriched cell population induces formation of midbrain structures, consistent with FGF8-organizing capability.
Collapse
Affiliation(s)
- Nils Offen
- Stem Cell and Developmental Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Alina Filatova
- Stem Cell and Developmental Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Ulrike A Nuber
- Stem Cell and Developmental Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany.
| |
Collapse
|
4
|
Sampath Kumar A, Tian L, Bolondi A, Hernández AA, Stickels R, Kretzmer H, Murray E, Wittler L, Walther M, Barakat G, Haut L, Elkabetz Y, Macosko EZ, Guignard L, Chen F, Meissner A. Spatiotemporal transcriptomic maps of whole mouse embryos at the onset of organogenesis. Nat Genet 2023; 55:1176-1185. [PMID: 37414952 PMCID: PMC10335937 DOI: 10.1038/s41588-023-01435-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/25/2023] [Indexed: 07/08/2023]
Abstract
Spatiotemporal orchestration of gene expression is required for proper embryonic development. The use of single-cell technologies has begun to provide improved resolution of early regulatory dynamics, including detailed molecular definitions of most cell states during mouse embryogenesis. Here we used Slide-seq to build spatial transcriptomic maps of complete embryonic day (E) 8.5 and E9.0, and partial E9.5 embryos. To support their utility, we developed sc3D, a tool for reconstructing and exploring three-dimensional 'virtual embryos', which enables the quantitative investigation of regionalized gene expression patterns. Our measurements along the main embryonic axes of the developing neural tube revealed several previously unannotated genes with distinct spatial patterns. We also characterized the conflicting transcriptional identity of 'ectopic' neural tubes that emerge in Tbx6 mutant embryos. Taken together, we present an experimental and computational framework for the spatiotemporal investigation of whole embryonic structures and mutant phenotypes.
Collapse
Affiliation(s)
- Abhishek Sampath Kumar
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Luyi Tian
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adriano Bolondi
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Amèlia Aragonés Hernández
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Robert Stickels
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lars Wittler
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maria Walther
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Gabriel Barakat
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Leah Haut
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Yechiel Elkabetz
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Evan Z Macosko
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Léo Guignard
- Aix Marseille University, Toulon University, Centre National de la Recherche Scientifique, Laboratoire d'Informatique et Systèmes 7020, Turing Centre for Living Systems, Marseille, France
| | - Fei Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
5
|
Lohoff T, Ghazanfar S, Missarova A, Koulena N, Pierson N, Griffiths JA, Bardot ES, Eng CHL, Tyser RCV, Argelaguet R, Guibentif C, Srinivas S, Briscoe J, Simons BD, Hadjantonakis AK, Göttgens B, Reik W, Nichols J, Cai L, Marioni JC. Integration of spatial and single-cell transcriptomic data elucidates mouse organogenesis. Nat Biotechnol 2022; 40:74-85. [PMID: 34489600 PMCID: PMC8763645 DOI: 10.1038/s41587-021-01006-2] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Molecular profiling of single cells has advanced our knowledge of the molecular basis of development. However, current approaches mostly rely on dissociating cells from tissues, thereby losing the crucial spatial context of regulatory processes. Here, we apply an image-based single-cell transcriptomics method, sequential fluorescence in situ hybridization (seqFISH), to detect mRNAs for 387 target genes in tissue sections of mouse embryos at the 8-12 somite stage. By integrating spatial context and multiplexed transcriptional measurements with two single-cell transcriptome atlases, we characterize cell types across the embryo and demonstrate that spatially resolved expression of genes not profiled by seqFISH can be imputed. We use this high-resolution spatial map to characterize fundamental steps in the patterning of the midbrain-hindbrain boundary (MHB) and the developing gut tube. We uncover axes of cell differentiation that are not apparent from single-cell RNA-sequencing (scRNA-seq) data, such as early dorsal-ventral separation of esophageal and tracheal progenitor populations in the gut tube. Our method provides an approach for studying cell fate decisions in complex tissues and development.
Collapse
Affiliation(s)
- T Lohoff
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Epigenetics Programme, Babraham Institute, Cambridge, UK
| | - S Ghazanfar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - A Missarova
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - N Koulena
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - N Pierson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - J A Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Genomics Plc, Cambridge, UK
| | - E S Bardot
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - C-H L Eng
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - R C V Tyser
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - R Argelaguet
- Epigenetics Programme, Babraham Institute, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - C Guibentif
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - S Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - J Briscoe
- The Francis Crick Institute, London, UK
| | - B D Simons
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- The Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - A-K Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - B Göttgens
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - W Reik
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Epigenetics Programme, Babraham Institute, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| | - J Nichols
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - L Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - J C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| |
Collapse
|
6
|
Isthmin1, a secreted signaling protein, acts downstream of diverse embryonic patterning centers in development. Cell Tissue Res 2020; 383:987-1002. [PMID: 33367974 PMCID: PMC7960586 DOI: 10.1007/s00441-020-03318-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/06/2020] [Indexed: 11/25/2022]
Abstract
Extracellular signals play essential roles during embryonic patterning by providing positional information in a concentration-dependent manner, and many such signals, like Wnt, fibroblast growth factor (FGF), Hedgehog (Hh), and retinoic acid, act by being secreted into the extracellular space, thereby triggering receptor-mediated responses in other cells. Isthmin1 (ism1) is a secreted protein whose gene expression pattern coincides with that of early dorsal determinants, nodal ligand genes like sqt and cyc, and with fgf8 during various phases of zebrafish development. Ism1 functions in early embryonic patterning and development are poorly understood; however, it has recently been shown to interact with nodal pathway genes to control organ asymmetry in chicken. Here, we show that misexpression of ism1 deletion constructs disrupts embryonic patterning in zebrafish and exhibits genetic interactions with both Fgf and nodal signaling. Unlike Fgf and nodal pathway mutants, CRISPR/Cas9-engineered ism1 mutants did not show obvious developmental defects. Further, in vivo single molecule fluorescence correlation spectroscopy (FCCS) showed that Ism1 diffuses freely in the extra-cellular space, with a diffusion coefficient similar to that of Fgf8a; however, our measurements do not support direct molecular interactions between Ism1 and either nodal ligands or Fgf8a in the developing zebrafish embryo. Together, data from gain- and loss-of-function experiments suggest that zebrafish Ism1 plays a complex role in regulating extracellular signals during early embryonic development.
Collapse
|
7
|
Alwin Prem Anand A, Alvarez-Bolado G, Wizenmann A. MiR-9 and the Midbrain-Hindbrain Boundary: A Showcase for the Limited Functional Conservation and Regulatory Complexity of MicroRNAs. Front Cell Dev Biol 2020; 8:586158. [PMID: 33330463 PMCID: PMC7719755 DOI: 10.3389/fcell.2020.586158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/23/2020] [Indexed: 11/15/2022] Open
Abstract
MicroRNAs regulate gene expression at post-transcriptional levels. Some of them appear to regulate brain development and are involved in neurodevelopmental disorders. This has led to the suggestion that the role of microRNAs in neuronal development and function may be more central than previously appreciated. Here, we review the data about miR-9 function to depict the subtlety, complexity, flexibility and limited functional conservation of this essential developmental regulatory system. On this basis we propose that species-specific actions of miR-9 could underlie to a large degree species differences in brain size, shape and function.
Collapse
Affiliation(s)
- A Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Tuebingen, Germany
| | | | - Andrea Wizenmann
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
8
|
Veerapathiran S, Teh C, Zhu S, Kartigayen I, Korzh V, Matsudaira PT, Wohland T. Wnt3 distribution in the zebrafish brain is determined by expression, diffusion and multiple molecular interactions. eLife 2020; 9:e59489. [PMID: 33236989 PMCID: PMC7725503 DOI: 10.7554/elife.59489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
Wnt3 proteins are lipidated and glycosylated signaling molecules that play an important role in zebrafish neural patterning and brain development. However, the transport mechanism of lipid-modified Wnts through the hydrophilic extracellular environment for long-range action remains unresolved. Here we determine how Wnt3 accomplishes long-range distribution in the zebrafish brain. First, we characterize the Wnt3-producing source and Wnt3-receiving target regions. Subsequently, we analyze Wnt3 mobility at different length scales by fluorescence correlation spectroscopy and fluorescence recovery after photobleaching. We demonstrate that Wnt3 spreads extracellularly and interacts with heparan sulfate proteoglycans (HSPG). We then determine the binding affinity of Wnt3 to its receptor, Frizzled1 (Fzd1), using fluorescence cross-correlation spectroscopy and show that the co-receptor, low-density lipoprotein receptor-related protein 5 (Lrp5), is required for Wnt3-Fzd1 interaction. Our results are consistent with the extracellular distribution of Wnt3 by a diffusive mechanism that is modified by tissue morphology, interactions with HSPG, and Lrp5-mediated receptor binding, to regulate zebrafish brain development.
Collapse
Affiliation(s)
- Sapthaswaran Veerapathiran
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
| | - Cathleen Teh
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
| | - Shiwen Zhu
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
| | - Indira Kartigayen
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | - Paul T Matsudaira
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
| | - Thorsten Wohland
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Center for BioImaging Sciences, National University of SingaporeSingaporeSingapore
- Department of Chemistry, National University of SingaporeSingaporeSingapore
| |
Collapse
|
9
|
Kesavan G, Machate A, Hans S, Brand M. Cell-fate plasticity, adhesion and cell sorting complementarily establish a sharp midbrain-hindbrain boundary. Development 2020; 147:dev186882. [PMID: 32439756 DOI: 10.1242/dev.186882] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/30/2020] [Indexed: 01/22/2023]
Abstract
The formation and maintenance of sharp boundaries between groups of cells play a vital role during embryonic development as they serve to compartmentalize cells with similar fates. Some of these boundaries also act as organizers, with the ability to induce specific cell fates and morphogenesis in the surrounding cells. The midbrain-hindbrain boundary (MHB) is such an organizer: it acts as a lineage restriction boundary to prevent the intermingling of cells with different developmental fates. However, the mechanisms underlying the lineage restriction process remain unclear. Here, using novel fluorescent knock-in reporters, live imaging, Cre/lox-mediated lineage tracing, atomic force microscopy-based cell adhesion assays and mutant analysis, we analyze the process of lineage restriction at the MHB and provide mechanistic details. Specifically, we show that lineage restriction occurs by the end of gastrulation, and that the subsequent formation of sharp gene expression boundaries in the developing MHB occur through complementary mechanisms, i.e. cell-fate plasticity and cell sorting. Furthermore, we show that cell sorting at the MHB involves differential adhesion among midbrain and hindbrain cells that is mediated by N-cadherin and Eph-ephrin signaling.
Collapse
Affiliation(s)
- Gokul Kesavan
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Anja Machate
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| |
Collapse
|
10
|
Lekven AC, Lilie CJ, Gibbs HC, Green DG, Singh A, Yeh AT. Analysis of the wnt1 regulatory chromosomal landscape. Dev Genes Evol 2019; 229:43-52. [PMID: 30825002 DOI: 10.1007/s00427-019-00629-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022]
Abstract
One of the earliest patterning events in the vertebrate neural plate is the specification of mes/r1, the territory comprising the prospective mesencephalon and the first hindbrain rhombomere. Within mes/r1, an interface of gene expression defines the midbrain-hindbrain boundary (MHB), a lineage restriction that separates the mesencephalon and rhombencephalon. wnt1 is critical to mes/r1 development and functions within the MHB as a component of the MHB gene regulatory network (GRN). Despite its importance to these critical and early steps of vertebrate neurogenesis, little is known about the factors responsible for wnt1 transcriptional regulation. In the zebrafish, wnt1 and its neighboring paralog, wnt10b, are expressed in largely overlapping patterns, suggesting co-regulation. To understand wnt1 and wnt10b transcriptional control, we used a comparative genomics approach to identify relevant enhancers. We show that the wnt1-wnt10b locus contains multiple cis-regulatory elements that likely interact to generate the wnt1 and wnt10b expression patterns. Two of 11 conserved enhancers tested show activity restricted to the midbrain and MHB, an activity that is conserved in the distantly related spotted gar orthologous elements. Three non-conserved elements also play a likely role in wnt1 regulation. The identified enhancers display dynamic modes of chromatin accessibility, suggesting controlled deployment during embryogenesis. Our results suggest that the control of wnt1 and wnt10b expression is under complex regulation involving the interaction of multiple enhancers.
Collapse
Affiliation(s)
- Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA. .,Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA.
| | - Craig J Lilie
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - David G Green
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Avantika Singh
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| |
Collapse
|
11
|
Jung J, Udhaya Kumar S, Choi I, Huh TL, Rhee M. Znf76 is associated with development of the eyes, midbrain, MHB, and hindbrain in zebrafish embryos. Anim Cells Syst (Seoul) 2019; 23:26-31. [PMID: 30834156 PMCID: PMC6394295 DOI: 10.1080/19768354.2018.1557744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 11/24/2022] Open
Abstract
ZNF76 is a transcriptional repressor that targets the TATA-binding protein (TBP) and plays an essential role during brain development; however, its function during embryogenesis remains unclear. Here, we report the expression pattern and potential functions of znf76 in zebrafish embryos. Maternal transcripts of znf76 were detected at low levels in embryos at the 1-cell stage, with zygotic transcripts appearing at the sphere stage. At the bud stage, the distribution of znf76 transcripts was polarized to the anterior and posterior regions of the embryos, and znf76 transcripts were further restricted to the trigeminal placode and proctodeum posterior gut of the embryos at 18 h postfertilization (hpf). znf76 transcripts were localized to the midbrain–hindbrain boundary (MHB), hindbrain, and developing eyes at 24 hpf. Ectopic expression of znf76 with 5’-capped znf76 mRNA microinjected into embryos at the 1-cell stage caused phenotypic defects in the eyes, MHB, hindbrain, and spinal cord. Overexpression of znf76 resulted in a drastic reduction of pax2a, fgf8a, and rx1 transcripts in the optic stalk, MHB, and eyes, respectively. Taken together, these data indicate that Znf76 governs developmental processes in the MHB, hindbrain, and eyes in zebrafish embryos. We also discuss the Fgf8 signaling networks associated with the Znf76 function.
Collapse
Affiliation(s)
- Jangham Jung
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, South Korea
| | - S Udhaya Kumar
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, South Korea
| | - Issac Choi
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, South Korea
| | - Tea-Lin Huh
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea
| | - Myungchull Rhee
- Department of Bioscience and Biotechnology, Graduate School, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
12
|
Alwin Prem Anand A, Huber C, Asnet Mary J, Gallus N, Leucht C, Klafke R, Hirt B, Wizenmann A. Expression and function of microRNA-9 in the mid-hindbrain area of embryonic chick. BMC DEVELOPMENTAL BIOLOGY 2018; 18:3. [PMID: 29471810 PMCID: PMC5824543 DOI: 10.1186/s12861-017-0159-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Background MiR-9 is a small non-coding RNA that is highly conserved between species and primarily expressed in the central nervous system (CNS). It is known to influence proliferation and neuronal differentiation in the brain and spinal cord of different vertebrates. Different studies have pointed to regional and species-specific differences in the response of neural progenitors to miR-9. Methods In ovo and ex ovo electroporation was used to overexpress or reduce miR-9 followed by mRNA in situ hybridisation and immunofluorescent stainings to evaluate miR- expression and the effect of changed miR-9 expression. Results We have investigated the expression and function of miR-9 during early development of the mid-hindbrain region (MH) in chick. Our analysis reveals a closer relationship of chick miR-9 to mammalian miR-9 than to fish and a dynamic expression pattern in the chick neural tube. Early in development, miR-9 is diffusely expressed in the entire brain, bar the forebrain, and it becomes more restricted to specific areas of the CNS at later stages. MiR-9 overexpression at HH9–10 results in a reduction of FGF8 expression and premature neuronal differentiation in the mid-hindbrain boundary (MHB). Within the midbrain miR-9 does not cause premature neuronal differentiation it rather reduces proliferation in the midbrain. Conclusion Our findings indicate that miR-9 has regional specific effects in the developing mid-hindbrain region with a divergence of response of regional progenitors. Electronic supplementary material The online version of this article (10.1186/s12861-017-0159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany
| | - Carola Huber
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.,Robert-Bosch-Krankenhaus, Auerbachstraße 110, 70376, Stuttgart, Germany
| | - John Asnet Mary
- Department of Zoology, Fatima College, Madurai, Tamilnadu, 625018, India
| | - Nancy Gallus
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.,Department of Neurobiology, McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Christoph Leucht
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ruth Klafke
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Bernhard Hirt
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany
| | - Andrea Wizenmann
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.
| |
Collapse
|
13
|
Kesavan G, Hammer J, Hans S, Brand M. Targeted knock-in of CreER T2 in zebrafish using CRISPR/Cas9. Cell Tissue Res 2018; 372:41-50. [DOI: 10.1007/s00441-018-2798-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/16/2018] [Indexed: 12/31/2022]
|
14
|
Lansdon LA, Darbro BW, Petrin AL, Hulstrand AM, Standley JM, Brouillette RB, Long A, Mansilla MA, Cornell RA, Murray JC, Houston DW, Manak JR. Identification of Isthmin 1 as a Novel Clefting and Craniofacial Patterning Gene in Humans. Genetics 2018; 208:283-296. [PMID: 29162626 PMCID: PMC5753863 DOI: 10.1534/genetics.117.300535] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022] Open
Abstract
Orofacial clefts are one of the most common birth defects, affecting 1-2 per 1000 births, and have a complex etiology. High-resolution array-based comparative genomic hybridization has increased the ability to detect copy number variants (CNVs) that can be causative for complex diseases such as cleft lip and/or palate. Utilizing this technique on 97 nonsyndromic cleft lip and palate cases and 43 cases with cleft palate only, we identified a heterozygous deletion of Isthmin 1 in one affected case, as well as a deletion in a second case that removes putative 3' regulatory information. Isthmin 1 is a strong candidate for clefting, as it is expressed in orofacial structures derived from the first branchial arch and is also in the same "synexpression group" as fibroblast growth factor 8 and sprouty RTK signaling antagonist 1a and 2, all of which have been associated with clefting. CNVs affecting Isthmin 1 are exceedingly rare in control populations, and Isthmin 1 scores as a likely haploinsufficiency locus. Confirming its role in craniofacial development, knockdown or clustered randomly interspaced short palindromic repeats/Cas9-generated mutation of isthmin 1 in Xenopus laevis resulted in mild to severe craniofacial dysmorphologies, with several individuals presenting with median clefts. Moreover, knockdown of isthmin 1 produced decreased expression of LIM homeobox 8, itself a gene associated with clefting, in regions of the face that pattern the maxilla. Our study demonstrates a successful pipeline from CNV identification of a candidate gene to functional validation in a vertebrate model system, and reveals Isthmin 1 as both a new human clefting locus as well as a key craniofacial patterning gene.
Collapse
Affiliation(s)
- Lisa A Lansdon
- Department of Pediatrics
- Department of Biology
- Interdisciplinary Graduate Program in Genetics
| | - Benjamin W Darbro
- Department of Pediatrics
- Interdisciplinary Graduate Program in Genetics
| | - Aline L Petrin
- Department of Pediatrics
- College of Dentistry, University of Iowa, Iowa 52242 and
| | | | | | | | | | | | - Robert A Cornell
- Interdisciplinary Graduate Program in Genetics
- Department of Anatomy and Cell Biology, and
| | - Jeffrey C Murray
- Department of Pediatrics
- Department of Biology
- Department of Anatomy and Cell Biology, and
- Interdisciplinary Graduate Program in Genetics
- College of Dentistry, University of Iowa, Iowa 52242 and
| | | | - J Robert Manak
- Department of Pediatrics,
- Department of Biology
- Interdisciplinary Graduate Program in Genetics
| |
Collapse
|
15
|
Brafman D, Willert K. Wnt/β-catenin signaling during early vertebrate neural development. Dev Neurobiol 2017; 77:1239-1259. [PMID: 28799266 DOI: 10.1002/dneu.22517] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 12/29/2022]
Abstract
The vertebrate central nervous system (CNS) is comprised of vast number of distinct cell types arranged in a highly organized manner. This high degree of complexity is achieved by cellular communication, including direct cell-cell contact, cell-matrix interactions, and cell-growth factor signaling. Among the several developmental signals controlling the development of the CNS, Wnt proteins have emerged as particularly critical and, hence, have captivated the attention of many researchers. With Wnts' evolutionarily conserved function as primordial symmetry breaking signals, these proteins and their downstream effects are responsible for simultaneously establishing cellular diversity and tissue organization. With their expansive repertoire of secreted agonists and antagonists, cell surface receptors, signaling cascades and downstream biological effects, Wnts are ideally suited to control the complex processes underlying vertebrate neural development. In this review, we will describe the mechanisms by which Wnts exert their potent effects on cells and tissues and highlight the many roles of Wnt signaling during neural development, starting from the initial induction of the neural plate, the subsequent patterning along the embryonic axes, to the intricately organized structure of the CNS. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1239-1259, 2017.
Collapse
Affiliation(s)
- David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287
| | - Karl Willert
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, 92093-0695
| |
Collapse
|
16
|
Gibbs HC, Chang-Gonzalez A, Hwang W, Yeh AT, Lekven AC. Midbrain-Hindbrain Boundary Morphogenesis: At the Intersection of Wnt and Fgf Signaling. Front Neuroanat 2017; 11:64. [PMID: 28824384 PMCID: PMC5541008 DOI: 10.3389/fnana.2017.00064] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
A constriction in the neural tube at the junction of the midbrain and hindbrain is a conserved feature of vertebrate embryos. The constriction is a defining feature of the midbrain-hindbrain boundary (MHB), a signaling center that patterns the adjacent midbrain and rostral hindbrain and forms at the junction of two gene expression domains in the early neural plate: an anterior otx2/wnt1 positive domain and a posterior gbx/fgf8 positive domain. otx2 and gbx genes encode mutually repressive transcription factors that create a lineage restriction boundary at their expression interface. Wnt and Fgf genes form a mutually dependent feedback system that maintains their expression domains on the otx2 or gbx side of the boundary, respectively. Constriction morphogenesis occurs after these conserved gene expression domains are established and while their mutual interactions maintain their expression pattern; consequently, mutant studies in zebrafish have led to the suggestion that constriction morphogenesis should be considered a unique phase of MHB development. We analyzed MHB morphogenesis in fgf8 loss of function zebrafish embryos using a reporter driven by the conserved wnt1 enhancer to visualize anterior boundary cells. We found that fgf8 loss of function results in a re-activation of wnt1 reporter expression posterior to the boundary simultaneous with an inactivation of the wnt1 reporter in the anterior boundary cells, and that these events correlate with relaxation of the boundary constriction. In consideration of other results that correlate the boundary constriction with Wnt and Fgf expression, we propose that the maintenance of an active Wnt-Fgf feedback loop is a key factor in driving the morphogenesis of the MHB constriction.
Collapse
Affiliation(s)
- Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Ana Chang-Gonzalez
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Wonmuk Hwang
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States.,Department of Materials Science and Engineering, Texas A&M UniversityCollege Station, TX, United States.,School of Computational Sciences, Korea Institute for Advanced StudySeoul, South Korea
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Arne C Lekven
- Department of Biology, Texas A&M UniversityCollege Station, TX, United States
| |
Collapse
|
17
|
Brignani S, Pasterkamp RJ. Neuronal Subset-Specific Migration and Axonal Wiring Mechanisms in the Developing Midbrain Dopamine System. Front Neuroanat 2017; 11:55. [PMID: 28740464 PMCID: PMC5502286 DOI: 10.3389/fnana.2017.00055] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/20/2017] [Indexed: 01/01/2023] Open
Abstract
The midbrain dopamine (mDA) system is involved in the control of cognitive and motor behaviors, and is associated with several psychiatric and neurodegenerative diseases. mDA neurons receive diverse afferent inputs and establish efferent connections with many brain areas. Recent studies have unveiled a high level of molecular and cellular heterogeneity within the mDA system with specific subsets of mDA neurons displaying select molecular profiles and connectivity patterns. During mDA neuron development, molecular differences between mDA neuron subsets allow the establishment of subset-specific afferent and efferent connections and functional roles. In this review, we summarize and discuss recent work defining novel mDA neuron subsets based on specific molecular signatures. Then, molecular cues are highlighted that control mDA neuron migration during embryonic development and that facilitate the formation of selective patterns of efferent connections. The review focuses largely on studies that show differences in these mechanisms between different subsets of mDA neurons and for which in vivo data is available, and is concluded by a section that discusses open questions and provides directions for further research.
Collapse
Affiliation(s)
- Sara Brignani
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
| | - R J Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
| |
Collapse
|
18
|
Kesavan G, Chekuru A, Machate A, Brand M. CRISPR/Cas9-Mediated Zebrafish Knock-in as a Novel Strategy to Study Midbrain-Hindbrain Boundary Development. Front Neuroanat 2017; 11:52. [PMID: 28713249 PMCID: PMC5492657 DOI: 10.3389/fnana.2017.00052] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/19/2017] [Indexed: 11/13/2022] Open
Abstract
The midbrain-hindbrain boundary (MHB) acts as an organizer and controls the fate of neighboring cells to develop into either mesencephalic (midbrain) or metencephalic (hindbrain) cells by secreting signaling molecules like Wnt1 and Fgf8. The zebrafish is an excellent vertebrate model for studying MHB development due to the ease of gene manipulation and the possibility of following cellular dynamics and morphogenetic processes using live imaging. Currently, only very few reporter and/or Cre-driver lines are available to study gene expression at the MHB, hampering the understanding of MHB development, and traditional transgenic technologies using promoter/enhancer fragments or bacterial artificial chromosome (BAC)-mediated transgenesis often do not faithfully recapitulate endogenous expression patterns. In contrast, CRISPR/Cas9-mediated genome editing technology now provides a great opportunity to efficiently knock-in or knock-out genes. We have generated four CRISPR/Cas9-based knock-in fluorescent reporter lines for two crucial genes involved in MHB development, namely otx2 and pax2a. The coding sequences of the reporters were knocked-in upstream of the corresponding ATG and are, thus, under the control of the endogenous promoter/enhancer elements. Interestingly, this strategy does not disturb endogenous gene expression. Using the fast maturing fluorescent protein reporter, Venus, enabled us to follow MHB development using cell tracking and live imaging. In addition, we show that these reporter lines label various neuronal and glial cell types in the adult zebrafish brain, making them highly suitable for investigating embryonic and adult midbrain, hindbrain, and MHB development.
Collapse
Affiliation(s)
- Gokul Kesavan
- Biotechnology Center and DFG-Research Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| | - Avinash Chekuru
- Biotechnology Center and DFG-Research Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| | - Anja Machate
- Biotechnology Center and DFG-Research Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| | - Michael Brand
- Biotechnology Center and DFG-Research Center for Regenerative Therapies Dresden, Technische Universität DresdenDresden, Germany
| |
Collapse
|
19
|
Fabian P, Pantzartzi CN, Kozmikova I, Kozmik Z. vox homeobox gene: a novel regulator of midbrain-hindbrain boundary development in medaka fish? Dev Genes Evol 2016; 226:99-107. [PMID: 26965282 DOI: 10.1007/s00427-016-0533-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/17/2016] [Indexed: 11/27/2022]
Abstract
The midbrain-hindbrain boundary (MHB) is one of the key organizing centers of the vertebrate central nervous system (CNS). Its patterning is governed by a well-described gene regulatory network (GRN) involving several transcription factors, namely, pax, gbx, en, and otx, together with signaling molecules of the Wnt and Fgf families. Here, we describe the onset of these markers in Oryzias latipes (medaka) early brain development in comparison to previously known zebrafish expression patterns. Moreover, we show for the first time that vox, a member of the vent gene family, is expressed in the developing neural tube similarly to CNS markers. Overexpression of vox leads to profound changes in the gene expression patterns of individual components of MHB-specific GRN, most notably of fgf8, a crucial organizer molecule of MHB. Our data suggest that genes from the vent family, in addition to their crucial role in body axis formation, may play a role in regionalization of vertebrate CNS.
Collapse
Affiliation(s)
- Peter Fabian
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, Czech Republic
| | - Chrysoula N Pantzartzi
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, Czech Republic
| | - Iryna Kozmikova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, Czech Republic
| | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, Czech Republic.
| |
Collapse
|
20
|
Aguillon R, Blader P, Batut J. Patterning, morphogenesis, and neurogenesis of zebrafish cranial sensory placodes. Methods Cell Biol 2016; 134:33-67. [PMID: 27312490 DOI: 10.1016/bs.mcb.2016.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Peripheral sensory organs and ganglia found in the vertebrate head arise during embryonic development from distinct ectodermal thickenings, called cranial sensory placodes (adenohypophyseal, olfactory, lens, trigeminal, epibranchial, and otic). A series of patterning events leads to the establishment of these placodes. Subsequently, these placodes undergo specific morphogenetic movements and cell-type specification in order to shape the final placodal derivatives and to produce differentiated cell types necessary for their function. In this chapter, we will focus on recent studies in the zebrafish that have advanced our understanding of cranial sensory placode development. We will summarize the signaling events and their molecular effectors guiding the formation of the so-called preplacodal region, and the subsequent subdivision of this region along the anteroposterior axis that gives rise to specific placode identities as well as those controlling morphogenesis and neurogenesis. Finally, we will highlight the approaches used in zebrafish that have been established to precisely label cell populations, to follow their development, and/or to characterize cell fates within a specific placode.
Collapse
Affiliation(s)
- R Aguillon
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - P Blader
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - J Batut
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
21
|
Tikhonov M, Little SC, Gregor T. Only accessible information is useful: insights from gradient-mediated patterning. ROYAL SOCIETY OPEN SCIENCE 2015; 2:150486. [PMID: 26716005 PMCID: PMC4680620 DOI: 10.1098/rsos.150486] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/29/2015] [Indexed: 06/01/2023]
Abstract
Information theory is gaining popularity as a tool to characterize performance of biological systems. However, information is commonly quantified without reference to whether or how a system could extract and use it; as a result, information-theoretic quantities are easily misinterpreted. Here, we take the example of pattern-forming developmental systems which are commonly structured as cascades of sequential gene expression steps. Such a multi-tiered structure appears to constitute sub-optimal use of the positional information provided by the input morphogen because noise is added at each tier. However, one must distinguish between the total information in a morphogen and information that can be usefully extracted and interpreted by downstream elements. We demonstrate that quantifying the information that is accessible to the system naturally explains the prevalence of multi-tiered network architectures as a consequence of the noise inherent to the control of gene expression. We support our argument with empirical observations from patterning along the major body axis of the fruit fly embryo. We use this example to highlight the limitations of the standard information-theoretic characterization of biological signalling, which are frequently de-emphasized, and illustrate how they can be resolved.
Collapse
Affiliation(s)
- Mikhail Tikhonov
- Joseph Henry Laboratories of Physics, Princeton University, Princeton, NJ 08544, USA
- Harvard Center of Mathematical Sciences and Applications, Harvard University, Cambridge, MA 02138, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Kavli Institute for Bionano Science and Technology, Harvard University, Cambridge, MA 02138, USA
| | - Shawn C. Little
- Joseph Henry Laboratories of Physics, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Thomas Gregor
- Joseph Henry Laboratories of Physics, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
22
|
Abdul-Wajid S, Morales-Diaz H, Khairallah SM, Smith WC. T-type Calcium Channel Regulation of Neural Tube Closure and EphrinA/EPHA Expression. Cell Rep 2015; 13:829-839. [PMID: 26489462 DOI: 10.1016/j.celrep.2015.09.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/06/2015] [Accepted: 09/11/2015] [Indexed: 10/22/2022] Open
Abstract
A major class of human birth defects arise from aberrations during neural tube closure (NTC). We report on a NTC signaling pathway requiring T-type calcium channels (TTCCs) that is conserved between primitive chordates (Ciona) and Xenopus. With loss of TTCCs, there is a failure to seal the anterior neural folds. Accompanying loss of TTCCs is an upregulation of EphrinA effectors. Ephrin signaling is known to be important in NTC, and ephrins can affect both cell adhesion and repulsion. In Ciona, ephrinA-d expression is downregulated at the end of neurulation, whereas, with loss of TTCC, ephrinA-d remains elevated. Accordingly, overexpression of ephrinA-d phenocopied TTCC loss of function, while overexpression of a dominant-negative Ephrin receptor was able to rescue NTC in a Ciona TTCC mutant. We hypothesize that signaling through TTCCs is necessary for proper anterior NTC through downregulation of ephrins, and possibly elimination of a repulsive signal.
Collapse
Affiliation(s)
- Sarah Abdul-Wajid
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Heidi Morales-Diaz
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Stephanie M Khairallah
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - William C Smith
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
23
|
Adutwum-Ofosu KK, Magnani D, Theil T, Price DJ, Fotaki V. The molecular and cellular signatures of the mouse eminentia thalami support its role as a signalling centre in the developing forebrain. Brain Struct Funct 2015; 221:3709-27. [PMID: 26459142 PMCID: PMC5009181 DOI: 10.1007/s00429-015-1127-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022]
Abstract
The mammalian eminentia thalami (EmT) (or thalamic eminence) is an embryonic forebrain structure of unknown function. Here, we examined the molecular and cellular properties of the mouse EmT. We first studied mRNA expression of signalling molecules and found that the EmT is a structure, rich in expression of secreted factors, with Wnts being the most abundantly detected. We then examined whether EmT tissue could induce cell fate changes when grafted ectopically. For this, we transplanted EmT tissue from a tau-GFP mouse to the ventral telencephalon of a wild type host, a telencephalic region where Wnt signalling is not normally active but which we showed in culture experiments is competent to respond to Wnts. We observed that the EmT was able to induce in adjacent ventral telencephalic cells ectopic expression of Lef1, a transcriptional activator and a target gene of the Wnt/β-catenin pathway. These Lef1-positive;GFP-negative cells expressed the telencephalic marker Foxg1 but not Ascl1, which is normally expressed by ventral telencephalic cells. These results suggest that the EmT has the capacity to activate Wnt/β-catenin signalling in the ventral telencephalon and to suppress ventral telencephalic gene expression. Altogether, our data support a role of the EmT as a signalling centre in the developing mouse forebrain.
Collapse
Affiliation(s)
- Kevin Kofi Adutwum-Ofosu
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Department of Anatomy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Dario Magnani
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Thomas Theil
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - David J Price
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Vassiliki Fotaki
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
24
|
Dyer C, Blanc E, Stanley RJ, Knight RD. Dissecting the role of Wnt signaling and its interactions with FGF signaling during midbrain neurogenesis. NEUROGENESIS 2015; 2:e1057313. [PMID: 27606327 PMCID: PMC4973611 DOI: 10.1080/23262133.2015.1057313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/07/2015] [Accepted: 05/27/2015] [Indexed: 11/14/2022]
Abstract
Interactions between FGF and Wnt/ bcat signaling control development of the midbrain. The nature of this interaction and how these regulate patterning, growth and differentiation is less clear, as it has not been possible to temporally dissect the effects of one pathway relative to the other. We have employed pharmacological and genetic tools to probe the temporal and spatial roles of FGF and Wnt in controlling the specification of early midbrain neurons. We identify a β-catenin (bcat) independent role for GSK-3 in modulating FGF activity and hence neuronal patterning. This function is complicated by an overlap with bcat-dependent regulation of FGF signaling, through the regulation of sprouty4. Additionally we reveal how attenuation of Axin protein function can promote fluctuating levels of bcat activity that are dependent on FGF activity. This highlights the complex nature of the interactions between FGF and Wnt/ bcat and reveals that they act at multiple levels to control each others activity in the midbrain.
Collapse
Affiliation(s)
- Carlene Dyer
- Craniofacial Development and Stem Cell Biology; King's College London ; London, UK
| | - Eric Blanc
- MRC Centre for Developmental Neurobiology; King's College London ; London, UK
| | - Rob J Stanley
- Department of Cell and Developmental Biology; University College London; London, UK; CoMPLEX; University College London; London, UK
| | - Robert D Knight
- Craniofacial Development and Stem Cell Biology; King's College London ; London, UK
| |
Collapse
|
25
|
Torres-Paz J, Whitlock KE. Olfactory sensory system develops from coordinated movements within the neural plate. Dev Dyn 2014; 243:1619-31. [PMID: 25255735 DOI: 10.1002/dvdy.24194] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 09/14/2014] [Accepted: 09/15/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The peripheral olfactory sensory system arises from morphologically identifiable structures called placodes. Placodes are relatively late developing structures, evident only well after the initiation of somitogenesis. Placodes are generally described as being induced from the ectoderm suggesting that their development is separate from the coordinated cell movements generating the central nervous system. RESULTS With the advent of modern techniques it is possible to follow the development of the neurectoderm giving rise to the anterior neural tube, including the olfactory placodes. The cell movements giving rise to the optic cup are coordinated with those generating the olfactory placodes and adjacent telencephalon. The formation of the basal lamina separating the placode from the neural tube is coincident with the anterior migration of cranial neural crest. CONCLUSIONS Olfactory placodes are transient morphological structures arising from a continuous sheet of neurectoderm that gives rise to the peripheral and central nervous system. This field of cells is specified at the end of gastrulation and not secondarily induced from ectoderm. The separation of olfactory placodes and telencephalon occurs through complex cell movements within the developing neural plate similar to that observed for the developing optic cup.
Collapse
Affiliation(s)
- Jorge Torres-Paz
- Centro Interdisciplinario de Neurociencia de Valparaiso, Instituto de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile
| | | |
Collapse
|
26
|
Osório L, Wu X, Zhou Z. Distinct spatiotemporal expression of ISM1 during mouse and chick development. Cell Cycle 2014; 13:1571-82. [PMID: 24675886 PMCID: PMC4050162 DOI: 10.4161/cc.28494] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 03/10/2014] [Indexed: 11/19/2022] Open
Abstract
Isthmin 1 (ISM1) constitutes the founder of a new family of secreted proteins characterized by the presence of 2 functional domains: thrombospondin type 1 repeat (TSR1) and adhesion-associated domain in MUC4 and other proteins (AMOP). ISM1 was identified in the frog embryo as a member of the FGF8 synexpression group due to its expression in the brain midbrain-hindbrain boundary (MHB) or isthmus. In zebrafish, ISM1 was described as a WNT- and NODAL-regulated gene. The function of ISM1 remains largely elusive. So far, ISM1 has been described as an angiogenesis inhibitor that has a dual function in endothelial cell survival and cell death. For a better understanding of ISM1 function, we examined its spatiotemporal distribution in mouse and chick using RT-PCR, ISH, and IHC analyses. In the mouse, ISM1 transcripts are found in tissues such as the anterior mesendoderm, paraxial and lateral plate mesoderm, MHB and trunk neural tube, as well as in the somites and dermomyotome. In the newborn and adult, ISM1 is prominently expressed in the lung and brain. In addition to its putative role during embryonic and postnatal development, ISM1 may also be important for organ homeostasis in the adult. In the chick embryo, ISM1 transcripts are strongly detected in the ear, eye, and spinal cord primordia. Remarkable differences in ISM1 spatiotemporal expression were found during mouse and chick development, despite the high homology of ISM1 orthologs in these species.
Collapse
Affiliation(s)
- Liliana Osório
- Department of Biochemistry; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong, China
- Shenzhen Institute of Research and Innovation; The University of Hong Kong; Shenzhen, China
| | - Xuewei Wu
- Department of Biochemistry; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong, China
- Shenzhen Institute of Research and Innovation; The University of Hong Kong; Shenzhen, China
| | - Zhongjun Zhou
- Department of Biochemistry; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong, China
- Shenzhen Institute of Research and Innovation; The University of Hong Kong; Shenzhen, China
| |
Collapse
|
27
|
Luz M, Spannl-Müller S, Özhan G, Kagermeier-Schenk B, Rhinn M, Weidinger G, Brand M. Dynamic association with donor cell filopodia and lipid-modification are essential features of Wnt8a during patterning of the zebrafish neuroectoderm. PLoS One 2014; 9:e84922. [PMID: 24427298 PMCID: PMC3888416 DOI: 10.1371/journal.pone.0084922] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/20/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Wnt proteins are conserved signaling molecules that regulate pattern formation during animal development. Many Wnt proteins are post-translationally modified by addition of lipid adducts. Wnt8a provides a crucial signal for patterning the anteroposterior axis of the developing neural plate in vertebrates. However, it is not clear how this protein propagates from its source, the blastoderm margin, to the target cells in the prospective neural plate, and how lipid-modifications might influence Wnt8a propagation and activity. RESULTS We have dynamically imaged biologically active, fluorescently tagged Wnt8a in living zebrafish embryos. We find that Wnt8a localizes to membrane-associated, punctate structures in live tissue. In Wnt8a expressing cells, these puncta are found on filopodial cellular processes, from where the protein can be released. In addition, Wnt8a is found colocalized with Frizzled receptor-containing clusters on signal receiving cells. Combining in vitro and in vivo assays, we compare the roles of conserved Wnt8a residues in cell and non-cell-autonomous signaling activity and secretion. Non-signaling Wnt8 variants show these residues can regulate Wnt8a distribution in producing cell membranes and filopodia as well as in the receiving tissue. CONCLUSIONS Together, our results show that Wnt8a forms dynamic clusters found on filopodial donor cell and on signal receiving cell membranes. Moreover, they demonstrate a differential requirement of conserved residues in Wnt8a protein for distribution in producing cells and receiving tissue and signaling activity during neuroectoderm patterning.
Collapse
Affiliation(s)
- Marta Luz
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Stephanie Spannl-Müller
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Günes Özhan
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | | | - Muriel Rhinn
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Gilbert Weidinger
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
28
|
Schmidt R, Strähle U, Scholpp S. Neurogenesis in zebrafish - from embryo to adult. Neural Dev 2013; 8:3. [PMID: 23433260 PMCID: PMC3598338 DOI: 10.1186/1749-8104-8-3] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/17/2013] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis in the developing central nervous system consists of the induction and proliferation of neural progenitor cells and their subsequent differentiation into mature neurons. External as well as internal cues orchestrate neurogenesis in a precise temporal and spatial way. In the last 20 years, the zebrafish has proven to be an excellent model organism to study neurogenesis in the embryo. Recently, this vertebrate has also become a model for the investigation of adult neurogenesis and neural regeneration. Here, we summarize the contributions of zebrafish in neural development and adult neurogenesis.
Collapse
Affiliation(s)
- Rebecca Schmidt
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76021, Karlsruhe, Germany
| | | | | |
Collapse
|
29
|
Pecchia L, Martin JL, Ragozzino A, Vanzanella C, Scognamiglio A, Mirarchi L, Morgan SP. User needs elicitation via analytic hierarchy process (AHP). A case study on a Computed Tomography (CT) scanner. BMC Med Inform Decis Mak 2013; 13:2. [PMID: 23289426 PMCID: PMC3545827 DOI: 10.1186/1472-6947-13-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 12/31/2012] [Indexed: 12/26/2022] Open
Abstract
Background The rigorous elicitation of user needs is a crucial step for both medical device design and purchasing. However, user needs elicitation is often based on qualitative methods whose findings can be difficult to integrate into medical decision-making. This paper describes the application of AHP to elicit user needs for a new CT scanner for use in a public hospital. Methods AHP was used to design a hierarchy of 12 needs for a new CT scanner, grouped into 4 homogenous categories, and to prepare a paper questionnaire to investigate the relative priorities of these. The questionnaire was completed by 5 senior clinicians working in a variety of clinical specialisations and departments in the same Italian public hospital. Results Although safety and performance were considered the most important issues, user needs changed according to clinical scenario. For elective surgery, the five most important needs were: spatial resolution, processing software, radiation dose, patient monitoring, and contrast medium. For emergency, the top five most important needs were: patient monitoring, radiation dose, contrast medium control, speed run, spatial resolution. Conclusions AHP effectively supported user need elicitation, helping to develop an analytic and intelligible framework of decision-making. User needs varied according to working scenario (elective versus emergency medicine) more than clinical specialization. This method should be considered by practitioners involved in decisions about new medical technology, whether that be during device design or before deciding whether to allocate budgets for new medical devices according to clinical functions or according to hospital department.
Collapse
Affiliation(s)
- Leandro Pecchia
- Electrical Systems and optics research division, Faculty of Engineering, University of Nottingham, NG7 2RD, Nottingham, UK.
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The cerebellum controls smooth and skillful movements and it is also involved in higher cognitive and emotional functions. The cerebellum is derived from the dorsal part of the anterior hindbrain and contains two groups of cerebellar neurons: glutamatergic and gamma-aminobutyric acid (GABA)ergic neurons. Purkinje cells are GABAergic and granule cells are glutamatergic. Granule and Purkinje cells receive input from outside of the cerebellum from mossy and climbing fibers. Genetic analysis of mice and zebrafish has revealed genetic cascades that control the development of the cerebellum and cerebellar neural circuits. During early neurogenesis, rostrocaudal patterning by intrinsic and extrinsic factors, such as Otx2, Gbx2 and Fgf8, plays an important role in the positioning and formation of the cerebellar primordium. The cerebellar glutamatergic neurons are derived from progenitors in the cerebellar rhombic lip, which express the proneural gene Atoh1. The GABAergic neurons are derived from progenitors in the ventricular zone, which express the proneural gene Ptf1a. The mossy and climbing fiber neurons originate from progenitors in the hindbrain rhombic lip that express Atoh1 or Ptf1a. Purkinje cells exhibit mediolateral compartmentalization determined on the birthdate of Purkinje cells, and linked to the precise neural circuitry formation. Recent studies have shown that anatomy and development of the cerebellum is conserved between mammals and bony fish (teleost species). In this review, we describe the development of cerebellar neurons and neural circuitry, and discuss their evolution by comparing developmental processes of mammalian and teleost cerebellum.
Collapse
Affiliation(s)
- Mitsuhiro Hashimoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi, 466-8550, Japan.
| | | |
Collapse
|
31
|
Crespo-Enriquez I, Partanen J, Martinez S, Echevarria D. Fgf8-related secondary organizers exert different polarizing planar instructions along the mouse anterior neural tube. PLoS One 2012; 7:e39977. [PMID: 22792203 PMCID: PMC3391221 DOI: 10.1371/journal.pone.0039977] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/30/2012] [Indexed: 12/11/2022] Open
Abstract
Early brain patterning depends on proper arrangement of positional information. This information is given by gradients of secreted signaling molecules (morphogens) detected by individual cells within the responding tissue, leading to specific fate decisions. Here we report that the morphogen FGF8 exerts initially a differential signal activity along the E9.5 mouse neural tube. We demonstrate that this polarizing activity codes by RAS-regulated ERK1/2 signaling and depends on the topographical location of the secondary organizers: the isthmic organizer (IsO) and the anterior neural ridge (anr) but not on zona limitans intrathalamica (zli). Our results suggest that Sprouty2, a negative modulator of RAS/ERK pathway, is important for regulating Fgf8 morphogenetic signal activity by controlling Fgf8-induced signaling pathways and positional information during early brain development.
Collapse
Affiliation(s)
- Ivan Crespo-Enriquez
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
| | - Juha Partanen
- Department of Biosciences, Division of Genetics, University of Helsinki, Helsinki, Finland
| | - Salvador Martinez
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
| | - Diego Echevarria
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
- * E-mail:
| |
Collapse
|
32
|
Mutual repression between Gbx2 and Otx2 in sensory placodes reveals a general mechanism for ectodermal patterning. Dev Biol 2012; 367:55-65. [PMID: 22564795 PMCID: PMC3384001 DOI: 10.1016/j.ydbio.2012.04.025] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 11/27/2022]
Abstract
In the vertebrate head, central and peripheral components of the sensory nervous system have different embryonic origins, the neural plate and sensory placodes. This raises the question of how they develop in register to form functional sense organs and sensory circuits. Here we show that mutual repression between the homeobox transcription factors Gbx2 and Otx2 patterns the placode territory by influencing regional identity and by segregating inner ear and trigeminal progenitors. Activation of Otx2 targets is necessary for anterior olfactory, lens and trigeminal character, while Gbx2 function is required for the formation of the posterior otic placode. Thus, like in the neural plate antagonistic interaction between Otx2 and Gbx2 establishes positional information thus providing a general mechanism for rostro-caudal patterning of the ectoderm. Our findings support the idea that the Otx/Gbx boundary has an ancient evolutionary origin to which different modules were recruited to specify cells of different fates.
Collapse
|
33
|
Cavodeassi F, Houart C. Brain regionalization: Of signaling centers and boundaries. Dev Neurobiol 2012; 72:218-33. [DOI: 10.1002/dneu.20938] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
34
|
Hochmann S, Kaslin J, Hans S, Weber A, Machate A, Geffarth M, Funk RHW, Brand M. Fgf signaling is required for photoreceptor maintenance in the adult zebrafish retina. PLoS One 2012; 7:e30365. [PMID: 22291943 PMCID: PMC3266925 DOI: 10.1371/journal.pone.0030365] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/19/2011] [Indexed: 11/19/2022] Open
Abstract
Fibroblast growth factors (Fgf) are secreted signaling molecules that have mitogenic, patterning, neurotrophic and angiogenic properties. Their importance during embryonic development in patterning and morphogenesis of the vertebrate eye is well known, but less is known about the role of Fgfs in the adult vertebrate retina. To address Fgf function in adult retina, we determined the spatial distribution of components of the Fgf signaling pathway in the adult zebrafish retina. We detected differential expression of Fgf receptors, ligands and downstream Fgf targets within specific retinal layers. Furthermore, we blocked Fgf signaling in the retina, by expressing a dominant negative variant of Fgf receptor 1 conditionally in transgenic animals. After blocking Fgf signaling we observe a fast and progressive photoreceptor degeneration and disorganization of retinal tissue, coupled with cell death in the outer nuclear layer. Following the degeneration of photoreceptors, a profound regeneration response is triggered that starts with proliferation in the inner nuclear layer. Ultimately, rod and cone photoreceptors are regenerated completely. Our study reveals the requirement of Fgf signaling to maintain photoreceptors and for proliferation during regeneration in the adult zebrafish retina.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Genetically Modified
- Cell Death/genetics
- Cell Death/physiology
- Cell Survival/genetics
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/physiology
- Gene Expression Regulation, Developmental
- Photoreceptor Cells, Vertebrate/metabolism
- Photoreceptor Cells, Vertebrate/physiology
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Receptors, Fibroblast Growth Factor/physiology
- Retina/cytology
- Retina/metabolism
- Retina/physiology
- Signal Transduction/genetics
- Signal Transduction/physiology
- Zebrafish/genetics
- Zebrafish/growth & development
- Zebrafish/metabolism
- Zebrafish/physiology
Collapse
Affiliation(s)
- Sarah Hochmann
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Jan Kaslin
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Stefan Hans
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Anke Weber
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Anja Machate
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Michaela Geffarth
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Richard H. W. Funk
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD), Biotechnology Center, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
35
|
Weisinger K, Kohl A, Kayam G, Monsonego-Ornan E, Sela-Donenfeld D. Expression of hindbrain boundary markers is regulated by FGF3. Biol Open 2011; 1:67-74. [PMID: 23213398 PMCID: PMC3507201 DOI: 10.1242/bio.2011032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Compartment boundaries act as organizing centers that segregate adjacent areas into domains of gene expression and regulation, and control their distinct fates via the secretion of signalling factors. During hindbrain development, a specialized cell-population forms boundaries between rhombomeres. These boundary cells demonstrate unique morphological properties and express multiple genes that differs them from intra-rhombomeric cells. Yet, little is known regarding the mechanisms that controls the expression or function of these boundary markers.Multiple components of the FGF signaling system, including ligands, receptors, downstream effectors as well as proteoglycans are shown to localize to boundary cells in the chick hindbrain. These patterns raise the possibility that FGF signaling plays a role in regulating boundary properties. We provide evidence to the role of FGF signaling, particularly the boundary-derived FGF3, in regulating the expression of multiple markers at hindbrain boundaries. These findings enable further characterization of the unique boundary-cell population, and expose a new function for FGFs as regulators of boundary-gene expression in the chick hindbrain.
Collapse
|
36
|
Fischer S, Filipek-Gorniok B, Ledin J. Zebrafish Ext2 is necessary for Fgf and Wnt signaling, but not for Hh signaling. BMC DEVELOPMENTAL BIOLOGY 2011; 11:53. [PMID: 21892940 PMCID: PMC3183004 DOI: 10.1186/1471-213x-11-53] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 09/05/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND Heparan sulfate (HS) biosynthesis is tightly regulated during vertebrate embryo development. However, potential roles for HS biosynthesis in regulating the function of paracrine signaling molecules that bind to HS are incompletely understood. RESULTS In this report we have studied Fgf, Wnt and Hedgehog (Hh) signaling in ext2 mutants, where heparan sulfate content is low. We found that Fgf targeted gene expression is reduced in ext2 mutants and that the remaining expression is readily inhibited by SU5402, an FGF receptor inhibitor. In the ext2 mutants, Fgf signaling is shown to be affected during nervous system development and reduction of Fgf ligands in the mutants affects tail development. Also, Wnt signaling is affected in the ext2 mutants, as shown by a stronger phenotype in ext2 mutants injected with morpholinos that partially block translation of Wnt11 or Wnt5b, compared to injected wild type embryos. In contrast, Hh dependent signaling is apparently unaffected in the ext2 mutants; Hh targeted gene expression is not reduced, the Hh inhibitor cyclopamine is not more affective in the mutants and Hh dependent cell differentiation in the retina and in the myotome are normal in ext2 mutants. In addition, no genetic interaction between ext2 and shha during development could be detected. CONCLUSION We conclude that ext2 is involved in Fgf and Wnt signaling but not in Hh signaling, revealing an unexpected specificity for ext2 in signaling pathways during embryonic development. Thus, our results support the hypothesis that regulation of heparan sulfate biosynthesis has distinct instructive functions for different signaling factors.
Collapse
Affiliation(s)
- Sabine Fischer
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | | |
Collapse
|
37
|
Transphyletic conservation of developmental regulatory state in animal evolution. Proc Natl Acad Sci U S A 2011; 108:14186-91. [PMID: 21844364 DOI: 10.1073/pnas.1109037108] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Specific regulatory states, i.e., sets of expressed transcription factors, define the gene expression capabilities of cells in animal development. Here we explore the functional significance of an unprecedented example of regulatory state conservation from the cnidarian Nematostella to Drosophila, sea urchin, fish, and mammals. Our probe is a deeply conserved cis-regulatory DNA module of the SRY-box B2 (soxB2), recognizable at the sequence level across many phyla. Transphyletic cis-regulatory DNA transfer experiments reveal that the plesiomorphic control function of this module may have been to respond to a regulatory state associated with neuronal differentiation. By introducing expression constructs driven by this module from any phyletic source into the genomes of diverse developing animals, we discover that the regulatory state to which it responds is used at different levels of the neurogenic developmental process, including patterning and development of the vertebrate forebrain and neurogenesis in the Drosophila optic lobe and brain. The regulatory state recognized by the conserved DNA sequence may have been redeployed to different levels of the developmental regulatory program during evolution of complex central nervous systems.
Collapse
|
38
|
Ganz J, Kaslin J, Hochmann S, Freudenreich D, Brand M. Heterogeneity and Fgf dependence of adult neural progenitors in the zebrafish telencephalon. Glia 2011; 58:1345-63. [PMID: 20607866 DOI: 10.1002/glia.21012] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adult telencephalic neurogenesis is a conserved trait of all vertebrates studied. It has been investigated in detail in rodents, but very little is known about the composition of neurogenic niches and the cellular nature of progenitors in nonmammalian vertebrates. To understand the components of the progenitor zones in the adult zebrafish telencephalon and the link between glial characteristics and progenitor state, we examined whether canonical glial markers are colocalized with proliferation markers. In the adult zebrafish telencephalon, we identify heterogeneous progenitors that reside in two distinct glial domains. We find that the glial composition of the progenitor zone is linked to its proliferative behavior. Analyzing both fast-cycling proliferating cells as well as slowly cycling progenitors, we find four distinct progenitor types characterized by differential expression of glial markers. Importantly, a significant proportion of progenitors do not display typical radial glia characteristics. By blocking or activating Fgf signaling by misexpression of a dominant negative Fgf-receptor 1 or Fgf8a, respectively, we find that ventral and dorsal progenitors in the telencephalon also differ in their requirement for Fgf signaling. Together with data on the expression of Fgf signaling components in the ventricular zone of the telencephalon, this suggests that Fgf signaling directly regulates proliferation of specific subsets of adult telencephalic progenitors in vivo. Taken together our results show that adult neural progenitor cells are heterogeneous with their respect to distribution into two distinct glial domains and their dependence upon Fgf signaling as a proliferative cue in the zebrafish telencephalon.
Collapse
Affiliation(s)
- Julia Ganz
- Biotechnology Center and Center for Regenerative Therapies Dresden, Dresden University of Technology, Tatzberg 47-49, 01307 Dresden, Germany
| | | | | | | | | |
Collapse
|
39
|
Pecchia L, Bath PA, Pendleton N, Bracale M. Analytic Hierarchy Process (AHP) for examining healthcare professionals' assessments of risk factors. The relative importance of risk factors for falls in community-dwelling older people. Methods Inf Med 2010; 50:435-44. [PMID: 21132218 DOI: 10.3414/me10-01-0028] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 10/25/2010] [Indexed: 11/09/2022]
Abstract
BACKGROUND A gap exists between evidence-based medicine and clinical-practice. Every day, healthcare professionals (HCPs) combine empirical evidence and subjective experience in order to maximize the effectiveness of interventions. Consequently, it is important to understand how HCPs interpret the research evidence and apply it in everyday practice. We focused on the prevention of falls, a common cause of injury-related morbidity and mortality in later life, for which there is a wide range of known risk factors. OBJECTIVES To use the Analytic Hierarchy Process (AHP) to investigate the opinions of HCPs in prioritizing risk factors for preventing falls. METHODS We used the AHP to develop a hierarchy of risk factors for falls based on the knowledge and experience of experts. We submitted electronic questionnaires via the web, in order to reach a wider number of respondents. With a web service, we pooled the results and weighted the coherence and the experience of respondents. RESULTS Overall, 232 respondents participated in the study: 32 in the technical pilot study, nine in the scientific pilot study and 191 respondents in the main study. We identified a hierarchy of 35 risk factors, organized in two categories and six sub-categories. CONCLUSIONS The hierarchy of risk factors provides further insights into clinicians' perceptions of risk factors for falls. This hierarchy helps understand the relative importance that clinicians place on risk factors for falls in older people and why evidence-based guidelines are not always followed. This information may be helpful in improving intervention programs and in understanding how clinicians prioritize multiple risk factors in individual patients. The AHP method allows the opinions of HCPs to be investigated, giving appropriate weight to their coherence, background and experience.
Collapse
Affiliation(s)
- L Pecchia
- Department of Biomedical, Electronic and Telecommunication Engineering, University Federico II of Naples, Via Claudio 21, 80125 Naples, Italy.
| | | | | | | |
Collapse
|
40
|
Gao FB. Context-dependent functions of specific microRNAs in neuronal development. Neural Dev 2010; 5:25. [PMID: 20920300 PMCID: PMC2958854 DOI: 10.1186/1749-8104-5-25] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 10/01/2010] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate multiple developmental processes at the post-transcriptional level. Recent rapid progresses have demonstrated critical roles for a number of miRNAs in neuronal development and function. In particular, miR-9 and miR-124 are specifically expressed in the mammalian nervous system, and their respective nucleotide sequences are 100% identical among many species. Yet, their expression patterns and mRNA targets are less conserved throughout evolution. As a consequence, these miRNAs exhibit diverse context-dependent functions in different aspects of neuronal development, ranging from early neurogenesis and neuronal differentiation to dendritic morphogenesis and synaptic plasticity. Some other neuronal miRNAs also exhibit context-dependent functions in development. Thus, post-transcriptional regulation of spatial and temporal expression levels of protein-coding genes by miRNAs contributes uniquely to the proper development and evolution of the complex nervous system.
Collapse
Affiliation(s)
- Fen-Biao Gao
- Department of Neurology and Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
41
|
Wong L, Weadick CJ, Kuo C, Chang BSW, Tropepe V. Duplicate dmbx1 genes regulate progenitor cell cycle and differentiation during zebrafish midbrain and retinal development. BMC DEVELOPMENTAL BIOLOGY 2010; 10:100. [PMID: 20860823 PMCID: PMC2954992 DOI: 10.1186/1471-213x-10-100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/22/2010] [Indexed: 01/03/2023]
Abstract
Background The Dmbx1 gene is important for the development of the midbrain and hindbrain, and mouse gene targeting experiments reveal that this gene is required for mediating postnatal and adult feeding behaviours. A single Dmbx1 gene exists in terrestrial vertebrate genomes, while teleost genomes have at least two paralogs. We compared the loss of function of the zebrafish dmbx1a and dmbx1b genes in order to gain insight into the molecular mechanism by which dmbx1 regulates neurogenesis, and to begin to understand why these duplicate genes have been retained in the zebrafish genome. Results Using gene knockdown experiments we examined the function of the dmbx1 gene paralogs in zebrafish, dmbx1a and dmbx1b in regulating neurogenesis in the developing retina and midbrain. Dose-dependent loss of dmbx1a and dmbx1b function causes a significant reduction in growth of the midbrain and retina that is evident between 48-72 hpf. We show that this phenotype is not due to patterning defects or persistent cell death, but rather a deficit in progenitor cell cycle exit and differentiation. Analyses of the morphant retina or anterior hindbrain indicate that paralogous function is partially diverged since loss of dmbx1a is more severe than loss of dmbx1b. Molecular evolutionary analyses of the Dmbx1 genes suggest that while this gene family is conservative in its evolution, there was a dramatic change in selective constraint after the duplication event that gave rise to the dmbx1a and dmbx1b gene families in teleost fish, suggestive of positive selection. Interestingly, in contrast to zebrafish dmbx1a, over expression of the mouse Dmbx1 gene does not functionally compensate for the zebrafish dmbx1a knockdown phenotype, while over expression of the dmbx1b gene only partially compensates for the dmbx1a knockdown phenotype. Conclusion Our data suggest that both zebrafish dmbx1a and dmbx1b genes are retained in the fish genome due to their requirement during midbrain and retinal neurogenesis, although their function is partially diverged. At the cellular level, Dmbx1 regulates cell cycle exit and differentiation of progenitor cells. The unexpected observation of putative post-duplication positive selection of teleost Dmbx1 genes, especially dmbx1a, and the differences in functionality between the mouse and zebrafish genes suggests that the teleost Dmbx1 genes may have evolved a diverged function in the regulation of neurogenesis.
Collapse
Affiliation(s)
- Loksum Wong
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
42
|
Abstract
For more than a decade, the zebrafish has proven to be an excellent model organism to investigate the mechanisms of neurogenesis during development. The often cited advantages, namely external development, genetic, and optical accessibility, have permitted direct examination and experimental manipulations of neurogenesis during development. Recent studies have begun to investigate adult neurogenesis, taking advantage of its widespread occurrence in the mature zebrafish brain to investigate the mechanisms underlying neural stem cell maintenance and recruitment. Here we provide a comprehensive overview of the tools and techniques available to study neurogenesis in zebrafish both during development and in adulthood. As useful resources, we provide tables of available molecular markers, transgenic, and mutant lines. We further provide optimized protocols for studying neurogenesis in the adult zebrafish brain, including in situ hybridization, immunohistochemistry, in vivo lipofection and electroporation methods to deliver expression constructs, administration of bromodeoxyuridine (BrdU), and finally slice cultures. These currently available tools have put zebrafish on par with other model organisms used to investigate neurogenesis.
Collapse
Affiliation(s)
- Prisca Chapouton
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | |
Collapse
|
43
|
Picker A, Cavodeassi F, Machate A, Bernauer S, Hans S, Abe G, Kawakami K, Wilson SW, Brand M. Dynamic coupling of pattern formation and morphogenesis in the developing vertebrate retina. PLoS Biol 2009; 7:e1000214. [PMID: 19823566 PMCID: PMC2751823 DOI: 10.1371/journal.pbio.1000214] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 08/28/2009] [Indexed: 12/25/2022] Open
Abstract
During embryonic development, pattern formation must be tightly synchronized with tissue morphogenesis to coordinate the establishment of the spatial identities of cells with their movements. In the vertebrate retina, patterning along the dorsal-ventral and nasal-temporal (anterior-posterior) axes is required for correct spatial representation in the retinotectal map. However, it is unknown how specification of axial cell positions in the retina occurs during the complex process of early eye morphogenesis. Studying zebrafish embryos, we show that morphogenetic tissue rearrangements during eye evagination result in progenitor cells in the nasal half of the retina primordium being brought into proximity to the sources of three fibroblast growth factors, Fgf8/3/24, outside the eye. Triple-mutant analysis shows that this combined Fgf signal fully controls nasal retina identity by regulating the nasal transcription factor Foxg1. Surprisingly, nasal-temporal axis specification occurs very early along the dorsal-ventral axis of the evaginating eye. By in vivo imaging GFP-tagged retinal progenitor cells, we find that subsequent eye morphogenesis requires gradual tissue compaction in the nasal half and directed cell movements into the temporal half of the retina. Balancing these processes drives the progressive alignment of the nasal-temporal retina axis with the anterior-posterior body axis and is controlled by a feed-forward effect of Fgf signaling on Foxg1-mediated cell cohesion. Thus, the mechanistic coupling and dynamic synchronization of tissue patterning with morphogenetic cell behavior through Fgf signaling leads to the graded allocation of cell positional identity in the eye, underlying retinotectal map formation.
Collapse
Affiliation(s)
- Alexander Picker
- Center of Regenerative Therapies Dresden, Biotechnology Center, Dresden University of Technology, Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chen Y, Mohammadi M, Flanagan JG. Graded levels of FGF protein span the midbrain and can instruct graded induction and repression of neural mapping labels. Neuron 2009; 62:773-80. [PMID: 19555646 DOI: 10.1016/j.neuron.2009.05.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 03/06/2009] [Accepted: 05/12/2009] [Indexed: 02/02/2023]
Abstract
Graded guidance labels are widely used in neural map formation, but it is not well understood which potential strategy leads to their graded expression. In midbrain tectal map development, FGFs can induce an entire midbrain, but their protein distribution is unclear, nor is it known whether they may act instructively to produce graded gene expression. Using a receptor-alkaline phosphatase fusion probe, we find a long-range posterior > anterior FGF protein gradient spanning the midbrain. Heparan sulfate proteoglycan (HSPG) is required for this gradient. To test whether graded FGF concentrations can instruct graded gene expression, a quantitative tectal explant assay was developed. Engrailed-2 and ephrin-As, normally in posterior > anterior tectal gradients, showed graded upregulation. Moreover, EphAs, normally in anterior > posterior countergradients, showed coordinately graded downregulation. These results provide a mechanism to establish graded mapping labels and more generally provide a developmental strategy to coordinately induce a structure and pattern its cell properties in gradients.
Collapse
Affiliation(s)
- Yao Chen
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
45
|
Apc1 is required for maintenance of local brain organizers and dorsal midbrain survival. Dev Biol 2009; 331:101-12. [PMID: 19397905 DOI: 10.1016/j.ydbio.2009.04.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/19/2009] [Accepted: 04/20/2009] [Indexed: 11/22/2022]
Abstract
The tumor suppressor Apc1 is an intracellular antagonist of the Wnt/beta-catenin pathway, which is vital for induction and patterning of the early vertebrate brain. However, its role in later brain development is less clear. Here, we examined the mechanisms underlying effects of an Apc1 zygotic-effect mutation on late brain development in zebrafish. Apc1 is required for maintenance of established brain subdivisions and control of local organizers such as the isthmic organizer (IsO). Caudal expansion of Fgf8 from IsO into the cerebellum is accompanied by hyperproliferation and abnormal cerebellar morphogenesis. Loss of apc1 results in reduced proliferation and apoptosis in the dorsal midbrain. Mosaic analysis shows that Apc is required cell-autonomously for maintenance of dorsal midbrain cell fate. The tectal phenotype occurs independently of Fgf8-mediated IsO function and is predominantly caused by stabilization of beta-catenin and subsequent hyperactivation of Wnt/beta-catenin signalling, which is mainly mediated through LEF1 activity. Chemical activation of the Wnt/beta-catenin in wild-type embryos during late brain maintenance stages phenocopies the IsO and tectal phenotypes of the apc mutants. These data demonstrate that Apc1-mediated restriction of Wnt/beta-catenin signalling is required for maintenance of local organizers and tectal integrity.
Collapse
|
46
|
Rhinn M, Lun K, Ahrendt R, Geffarth M, Brand M. Zebrafish gbx1 refines the midbrain-hindbrain boundary border and mediates the Wnt8 posteriorization signal. Neural Dev 2009; 4:12. [PMID: 19341460 PMCID: PMC2674439 DOI: 10.1186/1749-8104-4-12] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 04/02/2009] [Indexed: 12/27/2022] Open
Abstract
Background Studies in mouse, Xenopus and chicken have shown that Otx2 and Gbx2 expression domains are fundamental for positioning the midbrain-hindbrain boundary (MHB) organizer. Of the two zebrafish gbx genes, gbx1 is a likely candidate to participate in this event because its early expression is similar to that reported for Gbx2 in other species. Zebrafish gbx2, on the other hand, acts relatively late at the MHB. To investigate the function of zebrafish gbx1 within the early neural plate, we used a combination of gain- and loss-of-function experiments. Results We found that ectopic gbx1 expression in the anterior neural plate reduces forebrain and midbrain, represses otx2 expression and repositions the MHB to a more anterior position at the new gbx1/otx2 border. In the case of gbx1 loss-of-function, the initially robust otx2 domain shifts slightly posterior at a given stage (70% epiboly), as does MHB marker expression. We further found that ectopic juxtaposition of otx2 and gbx1 leads to ectopic activation of MHB markers fgf8, pax2.1 and eng2. This indicates that, in zebrafish, an interaction between otx2 and gbx1 determines the site of MHB development. Our work also highlights a novel requirement for gbx1 in hindbrain development. Using cell-tracing experiments, gbx1 was found to cell-autonomously transform anterior neural tissue into posterior. Previous studies have shown that gbx1 is a target of Wnt8 graded activity in the early neural plate. Consistent with this, we show that gbx1 can partially restore hindbrain patterning in cases of Wnt8 loss-of-function. We propose that in addition to its role at the MHB, gbx1 acts at the transcriptional level to mediate Wnt8 posteriorizing signals that pattern the developing hindbrain. Conclusion Our results provide evidence that zebrafish gbx1 is involved in positioning the MHB in the early neural plate by refining the otx2 expression domain. In addition to its role in MHB formation, we have shown that gbx1 is a novel mediator of Wnt8 signaling during hindbrain patterning.
Collapse
Affiliation(s)
- Muriel Rhinn
- Biotechnology Center, and Center for Regenerative Therapies Dresden, CRTD, Dresden University of Technology, Dresden, Germany.
| | | | | | | | | |
Collapse
|
47
|
Yamauchi K, Mizushima S, Tamada A, Yamamoto N, Takashima S, Murakami F. FGF8 signaling regulates growth of midbrain dopaminergic axons by inducing semaphorin 3F. J Neurosci 2009; 29:4044-55. [PMID: 19339600 PMCID: PMC6665371 DOI: 10.1523/jneurosci.4794-08.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 01/28/2009] [Accepted: 02/11/2009] [Indexed: 11/21/2022] Open
Abstract
Accumulating evidence indicates that signaling centers controlling the dorsoventral (DV) polarization of the neural tube, the roof plate and the floor plate, play crucial roles in axon guidance along the DV axis. However, the role of signaling centers regulating the rostrocaudal (RC) polarization of the neural tube in axon guidance along the RC axis remains unknown. Here, we show that a signaling center located at the midbrain-hindbrain boundary (MHB) regulates the rostrally directed growth of axons from midbrain dopaminergic neurons (mDANs). We found that beads soaked with fibroblast growth factor 8 (FGF8), a signaling molecule that mediates patterning activities of the MHB, repelled mDAN axons that extended through the diencephalon. This repulsion may be mediated by semaphorin 3F (sema3F) because (1) FGF8-soaked beads induced an increase in expression of sema3F, (2) sema3F expression in the midbrain was essentially abolished by the application of an FGF receptor tyrosine kinase inhibitor, and (3) mDAN axonal growth was also inhibited by sema3F. Furthermore, mDAN axons expressed a sema3F receptor, neuropilin-2 (nrp2), and the removal of nrp-2 by gene targeting caused caudal growth of mDAN axons. These results indicate that the MHB signaling center regulates the growth polarity of mDAN axons along the RC axis by inducing sema3F.
Collapse
Affiliation(s)
- Kenta Yamauchi
- Laboratory of Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Shigeki Mizushima
- Laboratory of Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Atsushi Tamada
- Division of Behavior and Neurobiology, National Institute for Basic Biology, Okazaki 444-8585, Japan, and
| | - Nobuhiko Yamamoto
- Laboratory of Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Seiji Takashima
- Department of Molecular Cardiology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Fujio Murakami
- Laboratory of Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
- Division of Behavior and Neurobiology, National Institute for Basic Biology, Okazaki 444-8585, Japan, and
| |
Collapse
|
48
|
Shima T, Znosko W, Tsang M. The characterization of a zebrafish mid-hindbrain mutant, mid-hindbrain gone (mgo). Dev Dyn 2009; 238:899-907. [DOI: 10.1002/dvdy.21916] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
49
|
Sela-Donenfeld D, Kayam G, Wilkinson DG. Boundary cells regulate a switch in the expression of FGF3 in hindbrain rhombomeres. BMC DEVELOPMENTAL BIOLOGY 2009; 9:16. [PMID: 19232109 PMCID: PMC2656489 DOI: 10.1186/1471-213x-9-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 02/20/2009] [Indexed: 11/18/2022]
Abstract
Background During formation of the vertebrate central nervous system, the hindbrain is organized into segmental units, called rhombomeres (r). These cell-lineage restricted segments are separated by a subpopulation of cells known as boundary cells. Boundary cells display distinct molecular and cellular properties such as an elongated shape, enriched extracellular matrix components and a reduced proliferation rate compared to intra-rhombomeric cells. However, little is known regarding their functions and the mechanisms that regulate their formation. Results Hindbrain boundary cells express several signaling molecules, such as FGF3, which at earlier developmental stages is transiently expressed in specific rhombomeres. We show that chick embryos that lack boundary cells due to overexpression of truncated EphA4 receptor in the hindbrain have continued segmental expression of FGF3 at stages when it is normally restricted to hindbrain boundaries. Furthermore, surgical ablation of the boundary between r3 and r4, or blocking of the contact of r4 with boundary cells, results in sustained FGF3 expression in this segment. Conclusion These findings suggest that boundary cells are required for the downregulation of segmental FGF3, presumably mediated by a soluble factor(s) that emanates from boundaries. We propose that this new function of boundary cells enables a switch in gene expression that may be required for stage-specific functions of FGF3 in the developing hindbrain.
Collapse
Affiliation(s)
- Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Hebrew University, The Robert H Smith Faculty of Agriculture, Food and Environment, Rehovot, Israel.
| | | | | |
Collapse
|
50
|
Kiecker C, Lumsden A. Recent advances in neural development. F1000 BIOLOGY REPORTS 2009; 1:1. [PMID: 20948677 PMCID: PMC3100780 DOI: 10.3410/b1-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A surprisingly small number of signalling pathways are used reiteratively during neural development, eliciting very different responses depending on the cellular context. Thus, the way a neural cell responds to a given signal is as important as the signal itself and this responsiveness, also called competence, changes with time. Here we describe recent advances in elucidating the signalling pathways that operate in brain development.
Collapse
Affiliation(s)
- Clemens Kiecker
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | | |
Collapse
|