1
|
Zhou W, Cai W, Li Y, Gao L, Liu X, Liu S, Lei J, Zhang J, Wang Y, Jiang Z, Wu X, Fan X, Li F, Zheng L, Yuan W. The Interaction Between the asb5a and asb5b Subtypes Jointly Regulates the L-R Asymmetrical Development of the Heart in Zebrafish. Int J Mol Sci 2025; 26:2765. [PMID: 40141403 PMCID: PMC11943173 DOI: 10.3390/ijms26062765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The asb5 gene, a member of the Asb protein subfamily characterized by six ankyrin repeat domains, is highly conserved and comprises two subtypes, asb5a and asb5b, in zebrafish. Our previous research has demonstrated that a deficiency of the asb5 gene significantly impairs early cardiac contractile function, highlighting its close relationship with heart development. Zebrafish asb5 expression was disrupted by both morpholino (MO) antisense oligomer-mediated knockdown and a CRISPR-Cas9 system. A high-throughput RNA-Seq analysis was used to analyze the possible molecular regulatory mechanism of asb5 gene deletion leading to left-right (L-R) asymmetry defects in the heart. Whole-mount in situ hybridization (WISH) was conducted to evaluate gene expression patterns of Nodal signaling components and the positions of heart organs. Heart looping was defective in zebrafish asb5 morphants. Rescue experiments in the asb5-deficiency group (inactivating both asb5a and asb5b) demonstrated that the injection of either asb5a-mRNA or asb5b-mRNA alone was insufficient to rectify the abnormal L-R asymmetry of the heart. In contrast, the simultaneous injection of both asb5a-mRNA and asb5b-mRNA successfully rescued the morphological phenotype. A high-throughput RNA-Seq analysis of embryos at 48 h post fertilization (hpf) revealed that numerous genes associated with L-R asymmetry exhibited expression imbalances in the asb5-deficiency group. WISH further confirmed that the expression of genes such as fli1a, acta1b, hand2, has2, prrx1a, notch1b, and foxa3 were upregulated, while the expression of mei2a and tal1 was downregulated. These results indicated that loss of the asb5 gene in zebrafish led to the disordered development of L-R asymmetry in the heart, resulting in an imbalance in the expression of genes associated with the regulation of L-R asymmetry. Subsequently, we examined the expression patterns of classical Nodal signaling pathway-related genes using WISH. The results showed that the midline barrier factor gene lefty1 was downregulated at early stages in the asb5-deficiency group, and the expression of spaw and lefty2, which are specific to the left lateral plate mesoderm (LPM), was disrupted. This study reveals that the two subtypes of the asb5 gene in zebrafish, asb5a and asb5b, interact and jointly regulate the establishment of early cardiac L-R asymmetry through the Nodal-spaw-lefty signaling pathway.
Collapse
Affiliation(s)
- Wanbang Zhou
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
- Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (J.L.); (J.Z.)
| | - Wanwan Cai
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
- Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (J.L.); (J.Z.)
| | - Yongqing Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Luoqing Gao
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Xin Liu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Siyuan Liu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Junrong Lei
- Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (J.L.); (J.Z.)
| | - Jisheng Zhang
- Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (J.L.); (J.Z.)
| | - Yuequn Wang
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Zhigang Jiang
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Xiushan Wu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Xiongwei Fan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Fang Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| | - Lan Zheng
- Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (J.L.); (J.Z.)
| | - Wuzhou Yuan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China; (W.Z.); (W.C.); (Y.L.); (L.G.); (X.L.); (S.L.); (Y.W.); (Z.J.); (X.W.); (X.F.); (F.L.)
| |
Collapse
|
2
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [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: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Vellutini BC, Martín-Durán JM, Børve A, Hejnol A. Combinatorial Wnt signaling landscape during brachiopod anteroposterior patterning. BMC Biol 2024; 22:212. [PMID: 39300453 PMCID: PMC11414264 DOI: 10.1186/s12915-024-01988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Wnt signaling pathways play crucial roles in animal development. They establish embryonic axes, specify cell fates, and regulate tissue morphogenesis from the early embryo to organogenesis. It is becoming increasingly recognized that these distinct developmental outcomes depend upon dynamic interactions between multiple ligands, receptors, antagonists, and other pathway modulators, consolidating the view that a combinatorial "code" controls the output of Wnt signaling. However, due to the lack of comprehensive analyses of Wnt components in several animal groups, it remains unclear if specific combinations always give rise to specific outcomes, and if these combinatorial patterns are conserved throughout evolution. RESULTS In this work, we investigate the combinatorial expression of Wnt signaling components during the axial patterning of the brachiopod Terebratalia transversa. We find that T. transversa has a conserved repertoire of ligands, receptors, and antagonists. These genes are expressed throughout embryogenesis but undergo significant upregulation during axial elongation. At this stage, Frizzled domains occupy broad regions across the body while Wnt domains are narrower and distributed in partially overlapping patches; antagonists are mostly restricted to the anterior end. Based on their combinatorial expression, we identify a series of unique transcriptional subregions along the anteroposterior axis that coincide with the different morphological subdivisions of the brachiopod larval body. When comparing these data across the animal phylogeny, we find that the expression of Frizzled genes is relatively conserved, whereas the expression of Wnt genes is more variable. CONCLUSIONS Our results suggest that the differential activation of Wnt signaling pathways may play a role in regionalizing the anteroposterior axis of brachiopod larvae. More generally, our analyses suggest that changes in the receptor context of Wnt ligands may act as a mechanism for the evolution and diversification of the metazoan body axis.
Collapse
Affiliation(s)
- Bruno C Vellutini
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway.
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307, Dresden, Germany.
| | - José M Martín-Durán
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, Fogg Building, London, E1 4NS, UK
| | - Aina Børve
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway
- Department of Biological Sciences, Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway
| | - Andreas Hejnol
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway.
- Department of Biological Sciences, Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway.
- Institute of Zoology and Evolutionary Research, Friedrich Schiller University Jena, Erbertstraße 1, 07743, Jena, Germany.
| |
Collapse
|
4
|
Liu Y, Kossack ME, McFaul ME, Christensen LN, Siebert S, Wyatt SR, Kamei CN, Horst S, Arroyo N, Drummond IA, Juliano CE, Draper BW. Single-cell transcriptome reveals insights into the development and function of the zebrafish ovary. eLife 2022; 11:e76014. [PMID: 35588359 PMCID: PMC9191896 DOI: 10.7554/elife.76014] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Zebrafish are an established research organism that has made many contributions to our understanding of vertebrate tissue and organ development, yet there are still significant gaps in our understanding of the genes that regulate gonad development, sex, and reproduction. Unlike the development of many organs, such as the brain and heart that form during the first few days of development, zebrafish gonads do not begin to form until the larval stage (≥5 days post-fertilization). Thus, forward genetic screens have identified very few genes required for gonad development. In addition, bulk RNA-sequencing studies that identify genes expressed in the gonads do not have the resolution necessary to define minor cell populations that may play significant roles in the development and function of these organs. To overcome these limitations, we have used single-cell RNA sequencing to determine the transcriptomes of cells isolated from juvenile zebrafish ovaries. This resulted in the profiles of 10,658 germ cells and 14,431 somatic cells. Our germ cell data represents all developmental stages from germline stem cells to early meiotic oocytes. Our somatic cell data represents all known somatic cell types, including follicle cells, theca cells, and ovarian stromal cells. Further analysis revealed an unexpected number of cell subpopulations within these broadly defined cell types. To further define their functional significance, we determined the location of these cell subpopulations within the ovary. Finally, we used gene knockout experiments to determine the roles of foxl2l and wnt9b for oocyte development and sex determination and/or differentiation, respectively. Our results reveal novel insights into zebrafish ovarian development and function, and the transcriptome profiles will provide a valuable resource for future studies.
Collapse
Affiliation(s)
- Yulong Liu
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Michelle E Kossack
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Matthew E McFaul
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Lana N Christensen
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Stefan Siebert
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Sydney R Wyatt
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Caramai N Kamei
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Samuel Horst
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Nayeli Arroyo
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Iain A Drummond
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Celina E Juliano
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| |
Collapse
|
5
|
Westphal M, Panza P, Kastenhuber E, Wehrle J, Driever W. Wnt/β-catenin signaling promotes neurogenesis in the diencephalospinal dopaminergic system of embryonic zebrafish. Sci Rep 2022; 12:1030. [PMID: 35046434 PMCID: PMC8770493 DOI: 10.1038/s41598-022-04833-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/28/2021] [Indexed: 12/21/2022] Open
Abstract
Wnt/β-catenin signaling contributes to patterning, proliferation, and differentiation throughout vertebrate neural development. Wnt/β-catenin signaling is important for mammalian midbrain dopaminergic neurogenesis, while little is known about its role in ventral forebrain dopaminergic development. Here, we focus on the A11-like, Otp-dependent diencephalospinal dopaminergic system in zebrafish. We show that Wnt ligands, receptors and extracellular antagonist genes are expressed in the vicinity of developing Otp-dependent dopaminergic neurons. Using transgenic Wnt/β-catenin-reporters, we found that Wnt/β-catenin signaling activity is absent from these dopaminergic neurons, but detected Wnt/β-catenin activity in cells adjacent to the caudal DC5/6 clusters of Otp-dependent dopaminergic neurons. Pharmacological manipulations of Wnt/β-catenin signaling activity, as well as heat-shock driven overexpression of Wnt agonists and antagonists, interfere with the development of DC5/6 dopaminergic neurons, such that Wnt/β-catenin activity positively correlates with their number. Wnt/β-catenin activity promoted dopaminergic development specifically at stages when DC5/6 dopaminergic progenitors are in a proliferative state. Our data suggest that Wnt/β-catenin signaling acts in a spatially and temporally restricted manner on proliferative dopaminergic progenitors in the hypothalamus to positively regulate the size of the dopaminergic neuron groups DC5 and DC6.
Collapse
Affiliation(s)
- Markus Westphal
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany.,CIBSS and BIOSS-Centres for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Paolo Panza
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany.,Department of Developmental Genetics, Max-Planck-Institute for Heart and Lung Research, Ludwigstraße 43, 61231, Bad Nauheim, Germany
| | - Edda Kastenhuber
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany.,Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Johanna Wehrle
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany.,CIBSS and BIOSS-Centres for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany. .,CIBSS and BIOSS-Centres for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
| |
Collapse
|
6
|
Jones WD, Mullins MC. Cell signaling pathways controlling an axis organizing center in the zebrafish. Curr Top Dev Biol 2022; 150:149-209. [DOI: 10.1016/bs.ctdb.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
7
|
Strain maps characterize the symmetry of convergence and extension patterns during zebrafish gastrulation. Sci Rep 2021; 11:19357. [PMID: 34588480 PMCID: PMC8481280 DOI: 10.1038/s41598-021-98233-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
During gastrulation of the zebrafish embryo, the cap of blastoderm cells organizes into the axial body plan of the embryo with left–right symmetry and head–tail, dorsal–ventral polarities. Our labs have been interested in the mechanics of early development and have investigated whether these large-scale cell movements can be described as tissue-level mechanical strain by a tectonics-based approach. The first step is to image the positions of all nuclei from mid-epiboly to early segmentation by digital sheet light microscopy, organize the surface of the embryo into multi-cell spherical domains, construct velocity fields from the movements of these domains and extract strain rate maps from the change in density of the domains. During gastrulation, tensile/expansive and compressive strains in the axial and equatorial directions are detected as anterior and posterior expansion along the anterior–posterior axis and medial–lateral compression across the dorsal–ventral axis and corresponds to the well characterized morphological movements of convergence and extension. Following gastrulation strain is represented by localized medial expansion at the onset of segmentation and anterior expansion at the onset of neurulation. In addition to linear strain, symmetric patterns of rotation/curl are first detected in the animal hemispheres at mid-epiboly and then the vegetal hemispheres by the end of gastrulation. In embryos treated with C59, a Wnt inhibitor that inhibits head and tail extension, the axial extension and vegetal curl are absent. By analysing the temporal sequence of large-scale movements, deformations across the embryo can be attributed to a combination of epiboly and dorsal convergence-extension.
Collapse
|
8
|
Ngernsombat C, Prattapong P, Larbcharoensub N, Khotthong K, Janvilisri T. WNT8B as an Independent Prognostic Marker for Nasopharyngeal Carcinoma. Curr Oncol 2021; 28:2529-2539. [PMID: 34287269 PMCID: PMC8293245 DOI: 10.3390/curroncol28040230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Members of the Wnt signaling pathway have been shown to play a role in nasopharyngeal carcinoma (NPC) progression. AIM The purpose of this study was to investigate WNT8B protein expression in NPC patients using tissue microarray (TMA) analysis and to evaluate its correlation with patient survival and clinical parameters. METHODS A total of 82 NPC cases, together with six normal nasopharyngeal tissue samples, were targeted to construct the TMA blocks. The WNT8B protein expression was evaluated by immunohistochemistry and its correlation to the clinicopathological features was investigated. RESULTS Sixty-two of 82 (75.6%) cases exhibited high WNT8B protein expression while 20/82 (24.4%) cases appeared to have low WNT8B expression. The univariate analysis revealed that systemic metastasis was associated with patient 5-year survival. The multivariate Cox proportional hazard regression analysis showed that WNT8B expression and systemic metastasis were significantly associated with the survival of NPC patients. Furthermore, there was no correlation found between the WNT8B protein expression and other clinicopathological parameters. CONCLUSION Our results suggest that the expression of WNT8B is associated with NPC patients' survival and could serve as an independent prognostic factor for NPC patients.
Collapse
Affiliation(s)
- Chawalit Ngernsombat
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (C.N.); (P.P.)
| | - Pongphol Prattapong
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (C.N.); (P.P.)
| | - Noppadol Larbcharoensub
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital Mahidol University, Bangkok 10400, Thailand;
| | | | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
9
|
Fuentes R, Tajer B, Kobayashi M, Pelliccia JL, Langdon Y, Abrams EW, Mullins MC. The maternal coordinate system: Molecular-genetics of embryonic axis formation and patterning in the zebrafish. Curr Top Dev Biol 2020; 140:341-389. [PMID: 32591080 DOI: 10.1016/bs.ctdb.2020.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Axis specification of the zebrafish embryo begins during oogenesis and relies on proper formation of well-defined cytoplasmic domains within the oocyte. Upon fertilization, maternally-regulated cytoplasmic flow and repositioning of dorsal determinants establish the coordinate system that will build the structure and developmental body plan of the embryo. Failure of specific genes that regulate the embryonic coordinate system leads to catastrophic loss of body structures. Here, we review the genetic principles of axis formation and discuss how maternal factors orchestrate axis patterning during zebrafish early embryogenesis. We focus on the molecular identity and functional contribution of genes controlling critical aspects of oogenesis, egg activation, blastula, and gastrula stages. We examine how polarized cytoplasmic domains form in the oocyte, which set off downstream events such as animal-vegetal polarity and germ line development. After gametes interact and form the zygote, cytoplasmic segregation drives the animal-directed reorganization of maternal determinants through calcium- and cell cycle-dependent signals. We also summarize how maternal genes control dorsoventral, anterior-posterior, mesendodermal, and left-right cell fate specification and how signaling pathways pattern these axes and tissues during early development to instruct the three-dimensional body plan. Advances in reverse genetics and phenotyping approaches in the zebrafish model are revealing positional patterning signatures at the single-cell level, thus enhancing our understanding of genotype-phenotype interactions in axis formation. Our emphasis is on the genetic interrogation of novel and specific maternal regulatory mechanisms of axis specification in the zebrafish.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Manami Kobayashi
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Jose L Pelliccia
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | | | - Elliott W Abrams
- Department of Biology, Purchase College, State University of New York, Harrison, NY, United States
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
10
|
Green DG, Whitener AE, Mohanty S, Mistretta B, Gunaratne P, Yeh AT, Lekven AC. Wnt signaling regulates neural plate patterning in distinct temporal phases with dynamic transcriptional outputs. Dev Biol 2020; 462:152-164. [PMID: 32243887 DOI: 10.1016/j.ydbio.2020.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 02/28/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
The process that partitions the nascent vertebrate central nervous system into forebrain, midbrain, hindbrain, and spinal cord after neural induction is of fundamental interest in developmental biology, and is known to be dependent on Wnt/β-catenin signaling at multiple steps. Neural induction specifies neural ectoderm with forebrain character that is subsequently posteriorized by graded Wnt signaling: embryological and mutant analyses have shown that progressively higher levels of Wnt signaling induce progressively more posterior fates. However, the mechanistic link between Wnt signaling and the molecular subdivision of the neural ectoderm into distinct domains in the anteroposterior (AP) axis is still not clear. To better understand how Wnt mediates neural AP patterning, we performed a temporal dissection of neural patterning in response to manipulations of Wnt signaling in zebrafish. We show that Wnt-mediated neural patterning in zebrafish can be divided into three phases: (I) a primary AP patterning phase, which occurs during gastrulation, (II) a mes/r1 (mesencephalon-rhombomere 1) specification and refinement phase, which occurs immediately after gastrulation, and (III) a midbrain-hindbrain boundary (MHB) morphogenesis phase, which occurs during segmentation stages. A major outcome of these Wnt signaling phases is the specification of the major compartment divisions of the developing brain: first the MHB, then the diencephalic-mesencephalic boundary (DMB). The specification of these lineage divisions depends upon the dynamic changes of gene transcription in response to Wnt signaling, which we show primarily involves transcriptional repression or indirect activation. We show that otx2b is directly repressed by Wnt signaling during primary AP patterning, but becomes resistant to Wnt-mediated repression during late gastrulation. Also during late gastrulation, Wnt signaling becomes both necessary and sufficient for expression of wnt8b, en2a, and her5 in mes/r1. We suggest that the change in otx2b response to Wnt regulation enables a transition to the mes/r1 phase of Wnt-mediated patterning, as it ensures that Wnts expressed in the midbrain and MHB do not suppress midbrain identity, and consequently reinforce formation of the DMB. These findings integrate important temporal elements into our spatial understanding of Wnt-mediated neural patterning and may serve as an important basis for a better understanding of neural patterning defects that have implications in human health.
Collapse
Affiliation(s)
- David G Green
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Amy E Whitener
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Saurav Mohanty
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Brandon Mistretta
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA.
| |
Collapse
|
11
|
Große A, Perner B, Naumann U, Englert C. Zebrafish Wtx is a negative regulator of Wnt signaling but is dispensable for embryonic development and organ homeostasis. Dev Dyn 2019; 248:866-881. [PMID: 31290212 DOI: 10.1002/dvdy.84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The X-chromosomally linked gene WTX is a human disease gene and a member of the AMER family. Mutations in WTX are found in Wilms tumor, a form of pediatric kidney cancer and in patients suffering from OSCS (Osteopathia striata with cranial sclerosis), a sclerosing bone disorder. Functional data suggest WTX to be an inhibitor of the Wnt/β-catenin signaling pathway. Deletion of Wtx in mouse leads to perinatal death, impeding the analysis of its physiological role. RESULTS To gain insights into the function of Wtx in development and homeostasis we have used zebrafish as a model and performed both knockdown and knockout studies using morpholinos and transcription activator-like effector nucleases (TALENs), respectively. Wtx knockdown led to increased Wnt activity and embryonic dorsalization. Also, wtx mutants showed a transient upregulation of Wnt target genes in the context of caudal fin regeneration. Surprisingly, however, wtx as well as wtx/amer2/amer3 triple mutants developed normally, were fertile and did not show any anomalies in organ maintenance. CONCLUSIONS Our data show that members of the zebrafish wtx/amer gene family, while sharing a partially overlapping expression pattern do not compensate for each other. This observation demonstrates a remarkable robustness during development and regeneration in zebrafish.
Collapse
Affiliation(s)
- Andreas Große
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Birgit Perner
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Uta Naumann
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Christoph Englert
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
12
|
Speer KF, Sommer A, Tajer B, Mullins MC, Klein PS, Lemmon MA. Non-acylated Wnts Can Promote Signaling. Cell Rep 2019; 26:875-883.e5. [PMID: 30673610 PMCID: PMC6429962 DOI: 10.1016/j.celrep.2018.12.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 10/27/2018] [Accepted: 12/26/2018] [Indexed: 12/14/2022] Open
Abstract
Wnts are a family of 19 extracellular ligands that regulate cell fate, proliferation, and migration during metazoan embryogenesis and throughout adulthood. Wnts are acylated post-translationally at a conserved serine and bind the extracellular cysteine-rich domain (CRD) of Frizzled (FZD) seven-pass transmembrane receptors. Although crystal structures suggest that acylation is essential for Wnt binding to FZDs, we show here that several Wnts can promote signaling in Xenopus laevis and Danio rerio embryos, as well as in an in vitro cell culture model, without acylation. The non-acylated Wnts are expressed at levels similar to wild-type counterparts and retain CRD binding. By contrast, we find that certain other Wnts do require acylation for biological activity in Xenopus embryos, although not necessarily for FZD binding. Our data argue that acylation dependence of Wnt activity is context specific. They further suggest that acylation may underlie aspects of ligand-receptor selectivity and/or control other aspects of Wnt function.
Collapse
Affiliation(s)
- Kelsey F Speer
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA; Department of Medicine (Hematology-Oncology), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5157, USA
| | - Anselm Sommer
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Tajer
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA
| | - Mary C Mullins
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA
| | - Peter S Klein
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA; Department of Medicine (Hematology-Oncology), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5157, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA.
| | - Mark A Lemmon
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6059, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
13
|
Mattes B, Dang Y, Greicius G, Kaufmann LT, Prunsche B, Rosenbauer J, Stegmaier J, Mikut R, Özbek S, Nienhaus GU, Schug A, Virshup DM, Scholpp S. Wnt/PCP controls spreading of Wnt/β-catenin signals by cytonemes in vertebrates. eLife 2018; 7:36953. [PMID: 30060804 PMCID: PMC6086664 DOI: 10.7554/elife.36953] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
Signaling filopodia, termed cytonemes, are dynamic actin-based membrane structures that regulate the exchange of signaling molecules and their receptors within tissues. However, how cytoneme formation is regulated remains unclear. Here, we show that Wnt/planar cell polarity (PCP) autocrine signaling controls the emergence of cytonemes, and that cytonemes subsequently control paracrine Wnt/β-catenin signal activation. Upon binding of the Wnt family member Wnt8a, the receptor tyrosine kinase Ror2 becomes activated. Ror2/PCP signaling leads to the induction of cytonemes, which mediate the transport of Wnt8a to neighboring cells. In the Wnt-receiving cells, Wnt8a on cytonemes triggers Wnt/β-catenin-dependent gene transcription and proliferation. We show that cytoneme-based Wnt transport operates in diverse processes, including zebrafish development, murine intestinal crypt and human cancer organoids, demonstrating that Wnt transport by cytonemes and its control via the Ror2 pathway is highly conserved in vertebrates. Communication helps the cells that make up tissues and organs to work together as a team. One way that cells share information with each other as tissues grow and develop is by exchanging signaling proteins. These interact with receptors on the surface of other cells; this causes the cell to change how it behaves. The Wnt family of signaling proteins orchestrate organ development. Wnt proteins influence which types of cells develop, how fast they divide, and how and when they move. Relatively few cells, or small groups of cells, in developing tissues produce Wnt proteins, while larger groups nearby respond to the signals. We do not fully understand how Wnt proteins travel between cells, but recent work revealed an unexpected mechanism – cells seem to hand-deliver their messages. Finger-like structures called cytonemes grow out of the cell membrane and carry Wnt proteins to their destination. If the cytonemes do not form properly the target cells do not behave correctly, which can lead to severe tissue malformation. Mattes et al. have now investigated how cytonemes form using a combination of state-of-the-art genetic and high-resolution imaging techniques. In initial experiments involving zebrafish cells that were grown in the laboratory, Mattes et al. found that the Wnt proteins kick start their own transport; before they travel to their destination, they act on the cells that made them. A Wnt protein called Wnt8a activates the receptor Ror2 on the surface of the signal-producing cell. Ror2 then triggers signals inside the cell that begin the assembly of the cytonemes. The more Ror2 is activated, the more cytonemes the cell makes, and the more Wnt signals it can send out. This mechanism operates in various tissues: Ror2 also controls the cytoneme transport process in living zebrafish embryos, the mouse intestine and human stomach tumors. This knowledge will help researchers to develop new ways to control Wnt signaling, which could help to produce new treatments for diseases ranging from cancers (for example in the stomach and bowel) to degenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Benjamin Mattes
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yonglong Dang
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gediminas Greicius
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Benedikt Prunsche
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Jakob Rosenbauer
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany
| | - Johannes Stegmaier
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Suat Özbek
- Centre of Organismal Studies, University of Heidelberg, Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Alexander Schug
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany.,Steinbuch Centre for Computing, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
14
|
Chen G, Zhang D, Zhang L, Feng G, Zhang B, Wu Y, Li W, Zhang Y, Hu B. RBM14 is indispensable for pluripotency maintenance and mesoderm development of mouse embryonic stem cells. Biochem Biophys Res Commun 2018; 501:259-265. [PMID: 29729270 DOI: 10.1016/j.bbrc.2018.04.231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
The pluripotency of embryonic stem cells (ESCs) is maintained by core pluripotency transcription factors, cofactors and several signaling pathways. RBM14 is a component of the para-speckle complex, which has been implicated in multiple important biological processes. The role of RBM14 in ESCs and lineage differentiation remains to be elucidated. In the present study, we provided evidence that RBM14 plays important roles in maintaining pluripotency and in the early differentiation of ESCs. RBM14 was demonstrated to be expressed in mouse embryonic stem cells (mESCs) and localized in the nucleus. RBM14 expression was depleted in mESCs using clustered regularly interspaced short palindromic repeats (CRISPR) technology. Our results also showed that RBM14 depletion altered the gene expression profiles of mESCs. In particular, pluripotency-associated genes and genes involved in the Wnt and TGF-β signaling pathways were downregulated in RBM14 knockout mESCs. Furthermore, RBM14 was found to be essential for mesoderm development in vitro and in vivo. The specific effects of RBM14 depletion were verified by conducting a rescue experiment. Our findings demonstrated that RBM14 not only plays an important role in maintaining the pluripotency of mESCs but is also indispensable for mesoderm development.
Collapse
Affiliation(s)
- Guilai Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Linlin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yihui Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
15
|
Jiang J, Tang S, Xia J, Wen J, Chen S, Shu X, Huen MSY, Deng Y. C9orf140, a novel Axin1-interacting protein, mediates the negative feedback loop of Wnt/β-catenin signaling. Oncogene 2018; 37:2992-3005. [PMID: 29531269 PMCID: PMC5978805 DOI: 10.1038/s41388-018-0166-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
Wnt/β-catenin signaling activity is maintained in homeostasis by an expanding list of molecular determinants. However, the molecular components and the regulatory mechanisms involved in its fine-tuning remain to be determined. Here, we identified C9orf140, a tumor-specific protein, as a novel Axin1-interacting protein by tandem-affinity purification and mass spectrometry. We further showed that C9orf140 is a negative regulator of Wnt/β-catenin signaling in cultured cells as well as in zebrafish embryos. It functions upstream of β-catenin, outcompetes PP2A for binding to Axin1, influences the balance between phosphorylation and de-phosphorylation of β-catenin, and ultimately compromises Wnt3A-induced β-catenin accumulation. Interestingly, Wnt-induced C9orf140 expression via β-catenin. We propose that C9orf140 mediates a negative feedback loop of Wnt/β-catenin signaling by interacting with Axin1. Our results advance the current understanding of the exquisite control of Wnt/β-catenin signaling cascade, and provide evidence of the new role of C9orf140.
Collapse
Affiliation(s)
- Jun Jiang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shulin Tang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jianhong Xia
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Shuang Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Xiaodong Shu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Michael S Y Huen
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
16
|
Yurt KK, Kaplan S. As a painkiller: a review of pre- and postnatal non-steroidal anti-inflammatory drug exposure effects on the nervous systems. Inflammopharmacology 2017; 26:15-28. [DOI: 10.1007/s10787-017-0434-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/10/2017] [Indexed: 11/29/2022]
|
17
|
Hino H, Nakanishi A, Seki R, Aoki T, Yamaha E, Kawahara A, Shimizu T, Hibi M. Roles of maternal wnt8a transcripts in axis formation in zebrafish. Dev Biol 2017; 434:96-107. [PMID: 29208373 DOI: 10.1016/j.ydbio.2017.11.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023]
Abstract
In early zebrafish development, the program for dorsal axis formation begins soon after fertilization. Previous studies suggested that dorsal determinants (DDs) localize to the vegetal pole, and are transported to the dorsal blastomeres in a microtubule-dependent manner. The DDs activate the canonical Wnt pathway and induce dorsal-specific genes that are required for dorsal axis formation. Among wnt-family genes, only the wnt8a mRNA is reported to localize to the vegetal pole in oocytes and to induce the dorsal axis, suggesting that Wnt8a is a candidate DD. Here, to reveal the roles of maternal wnt8a, we generated wnt8a mutants by transcription activator-like effector nucleases (TALENs), and established zygotic, maternal, and maternal zygotic wnt8a mutants by germ-line replacement. Zebrafish wnt8a has two open reading frames (ORF1 and ORF2) that are tandemly located in the genome. Although the zygotic ORF1 or ORF2 wnt8a mutants showed little or no axis-formation defects, the ORF1/2 compound mutants showed antero-dorsalized phenotypes, indicating that ORF1 and ORF2 have redundant roles in ventrolateral and posterior tissue formation. Unexpectedly, the maternal wnt8a ORF1/2 mutants showed no axis-formation defects. The maternal-zygotic wnt8a ORF1/2 mutants showed more severe antero-dorsalized phenotypes than the zygotic mutants. These results indicated that maternal wnt8a is dispensable for the initial dorsal determination, but cooperates with zygotic wnt8a for ventrolateral and posterior tissue formation. Finally, we re-examined the maternal wnt genes and found that Wnt6a is an alternative candidate DD.
Collapse
Affiliation(s)
- Hiromu Hino
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan; Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Akiko Nakanishi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Ryoko Seki
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Tsubasa Aoki
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Etsuro Yamaha
- Nanae Fresh Water Laboratory, Field Science Center for Northern Biosphere, Hokkaido University, Nanae, Kameda, Hokkaido 041-1105, Japan
| | - Atsuo Kawahara
- Laboratory for Developmental Biology, Center for Medical Education and Sciences, Graduate School of Medical Science, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Takashi Shimizu
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan; Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Masahiko Hibi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan; Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan.
| |
Collapse
|
18
|
Yuan Q, Zhao M, Tandon B, Maili L, Liu X, Zhang A, Baugh EH, Tran T, Silva RM, Hecht JT, Swindell EC, Wagner DS, Letra A. Role of WNT10A in failure of tooth development in humans and zebrafish. Mol Genet Genomic Med 2017; 5:730-741. [PMID: 29178643 PMCID: PMC5702573 DOI: 10.1002/mgg3.332] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Oligodontia is a severe form of tooth agenesis characterized by the absence of six or more permanent teeth. Oligodontia has complex etiology and variations in numerous genes have been suggested as causal for the condition. METHODS We applied whole-exome sequencing (WES) to identify the cause of oligodontia in a 9-year-old girl missing 11 permanent teeth. Protein modeling and functional analysis in zebrafish were also performed to understand the impact of identified variants on the phenotype. RESULTS We identified a novel compound heterozygous missense mutation in WNT10A (c.637G>A:p.Gly213Ser and c.1070C>T:p.Thr357Ile) as the likely cause of autosomal recessive oligodontia in the child. Affected residues are located in conserved regions and variants are predicted to be highly deleterious for potentially destabilizing the protein fold and inhibiting normal protein function. Functional studies in zebrafish embryos showed that wnt10a is expressed in the craniofacies at critical time points for tooth development, and that perturbations of wnt10a expression impaired normal tooth development and arrested tooth development at 5 days postfertilization (dpf). Furthermore, mRNA expression levels of additional tooth development genes were directly correlated with wnt10a expression; expression of msx1, dlx2b, eda, and axin2 was decreased upon wnt10a knockdown, and increased upon wnt10a overexpression. CONCLUSIONS Our results reveal a novel compound heterozygous variant in WNT10A as pathogenic for oligodontia, and demonstrate that perturbations of wnt10a expression in zebrafish may directly and/or indirectly affect tooth development recapitulating the agenesis phenotype observed in humans.
Collapse
Affiliation(s)
- Qiuping Yuan
- Department of PediatricsUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Pediatric Research CenterUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
| | - Min Zhao
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| | - Bhavna Tandon
- Department of BiosciencesRice UniversityHoustonTexas
| | - Lorena Maili
- Department of PediatricsUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Pediatric Research CenterUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
| | - Xiaoming Liu
- Department of Human GeneticsUniversity of Texas Health Science Center at Houston School of Public HealthHoustonTexas
| | - Anqi Zhang
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| | - Evan H. Baugh
- Department of BiologyNew York UniversityNew YorkNew York
| | - Tam Tran
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| | - Renato M. Silva
- Pediatric Research CenterUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
- Department of EndodonticsUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| | - Jacqueline T. Hecht
- Department of PediatricsUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Pediatric Research CenterUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| | - Eric C. Swindell
- University of Texas Graduate School of Biomedical Sciences at HoustonHoustonTexas77030
| | | | - Ariadne Letra
- Pediatric Research CenterUniversity of Texas Health Science Center at Houston Medical SchoolHoustonTexas
- Center for Craniofacial ResearchUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
- Department of Diagnostic and Biomedical SciencesUniversity of Texas Health Science Center at Houston School of DentistryHoustonTexas
| |
Collapse
|
19
|
Chin AM, Hill DR, Aurora M, Spence JR. Morphogenesis and maturation of the embryonic and postnatal intestine. Semin Cell Dev Biol 2017; 66:81-93. [PMID: 28161556 DOI: 10.1016/j.semcdb.2017.01.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
Abstract
The intestine is a vital organ responsible for nutrient absorption, bile and waste excretion, and a major site of host immunity. In order to keep up with daily demands, the intestine has evolved a mechanism to expand the absorptive surface area by undergoing a morphogenetic process to generate finger-like units called villi. These villi house specialized cell types critical for both absorbing nutrients from food, and for protecting the host from commensal and pathogenic microbes present in the adult gut. In this review, we will discuss mechanisms that coordinate intestinal development, growth, and maturation of the small intestine, starting from the formation of the early gut tube, through villus morphogenesis and into early postnatal life when the intestine must adapt to the acquisition of nutrients through food intake, and to interactions with microbes.
Collapse
Affiliation(s)
- Alana M Chin
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David R Hill
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Megan Aurora
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
20
|
Houston DW. Vertebrate Axial Patterning: From Egg to Asymmetry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:209-306. [PMID: 27975274 PMCID: PMC6550305 DOI: 10.1007/978-3-319-46095-6_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of the bilateral embryonic body axis from a symmetrical egg has been a long-standing question in developmental biology. Historical and modern experiments point to an initial symmetry-breaking event leading to localized Wnt and Nodal growth factor signaling and subsequent induction and formation of a self-regulating dorsal "organizer." This organizer forms at the site of notochord cell internalization and expresses primarily Bone Morphogenetic Protein (BMP) growth factor antagonists that establish a spatiotemporal gradient of BMP signaling across the embryo, directing initial cell differentiation and morphogenesis. Although the basics of this model have been known for some time, many of the molecular and cellular details have only recently been elucidated and the extent that these events remain conserved throughout vertebrate evolution remains unclear. This chapter summarizes historical perspectives as well as recent molecular and genetic advances regarding: (1) the mechanisms that regulate symmetry-breaking in the vertebrate egg and early embryo, (2) the pathways that are activated by these events, in particular the Wnt pathway, and the role of these pathways in the formation and function of the organizer, and (3) how these pathways also mediate anteroposterior patterning and axial morphogenesis. Emphasis is placed on comparative aspects of the egg-to-embryo transition across vertebrates and their evolution. The future prospects for work regarding self-organization and gene regulatory networks in the context of early axis formation are also discussed.
Collapse
Affiliation(s)
- Douglas W Houston
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA.
| |
Collapse
|
21
|
Scott CA, Marsden AN, Slusarski DC. Automated, high-throughput, in vivo analysis of visual function using the zebrafish. Dev Dyn 2016; 245:605-13. [PMID: 26890697 DOI: 10.1002/dvdy.24398] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Modern genomics has enabled the identification of an unprecedented number of genetic variants, which in many cases are extremely rare, associated with blinding disorders. A significant challenge will be determining the pathophysiology of each new variant. The Zebrafish is an excellent model for the study of inherited diseases of the eye. By 5 days post-fertilization (dpf), they have quantifiable behavioral responses to visual stimuli. However, visual behavior assays can take several hours to perform or can only be assessed one fish at a time. RESULTS To increase the throughput for vision assays, we used the Viewpoint Zebrabox to automate the visual startle response and created software, Visual Interrogation of Zebrafish Manipulations (VIZN), to automate data analysis. This process allows 96 Zebrafish larvae to be tested and resultant data to be analyzed in less than 35 minutes. We validated this system by disrupting function of a gene necessary for photoreceptor differentiation and observing decreased response to visual stimuli. CONCLUSIONS This automated method along with VIZN allows rapid, high-throughput, in vivo testing of Zebrafish's ability to respond to light/dark stimuli. This allows the rapid analysis of novel genes involved in visual function by morpholino, CRISPRS, or small-molecule drug screens. Developmental Dynamics 245:605-613, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Autumn N Marsden
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
22
|
Duncan RN, Panahi S, Piotrowski T, Dorsky RI. Identification of Wnt Genes Expressed in Neural Progenitor Zones during Zebrafish Brain Development. PLoS One 2015; 10:e0145810. [PMID: 26713625 PMCID: PMC4699909 DOI: 10.1371/journal.pone.0145810] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 12/09/2015] [Indexed: 12/31/2022] Open
Abstract
Wnt signaling regulates multiple aspects of vertebrate central nervous system (CNS) development, including neurogenesis. However, vertebrate genomes can contain up to 25 Wnt genes, the functions of which are poorly characterized partly due to redundancy in their expression. To identify candidate Wnt genes as candidate mediators of pathway activity in specific brain progenitor zones, we have performed a comprehensive expression analysis at three different stages during zebrafish development. Antisense RNA probes for 21 Wnt genes were generated from existing and newly synthesized cDNA clones and used for in situ hybridization on whole embryos and dissected brains. As in other species, we found that Wnt expression patterns in the embryonic zebrafish CNS are complex and often redundant. We observed that progenitor zones in the telencephalon, dorsal diencephalon, hypothalamus, midbrain, midbrain-hindbrain boundary, cerebellum and retina all express multiple Wnt genes. Our data identify 12 specific ligands that can now be tested using loss-of-function approaches.
Collapse
Affiliation(s)
- Robert N Duncan
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Samin Panahi
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
23
|
Thisse B, Thisse C. Formation of the vertebrate embryo: Moving beyond the Spemann organizer. Semin Cell Dev Biol 2015; 42:94-102. [PMID: 25999320 DOI: 10.1016/j.semcdb.2015.05.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/13/2015] [Indexed: 12/14/2022]
Abstract
During the course of their classic experiments, Hilde Mangold and Hans Spemann discovered that the dorsal blastopore lip of an amphibian gastrula was able to induce formation of a complete embryonic axis when transplanted into the ventral side of a host gastrula embryo. Since then, the inducing activity of the dorsal lip has been known as the Spemann or dorsal organizer. During the past 25 years, studies performed in a variety of species have led to the identification of molecular factors associated with the properties of this tissue. However, none of them is, by itself, able to induce formation of the main body axis from a population of naive pluripotent embryonic cells. Recently, experiments performed using the zebrafish (Danio rerio) revealed that the organizing activities present in the embryo are not restricted to the Spemann organizer but are distributed along the entire blastula/gastrula margin. These organizing activities result from the interaction between two opposing gradients of morphogens, BMP and Nodal, that are the primary signals that trigger the cascade of developmental events leading to the organization of the embryo. These studies mark the end of the era during which developmental biologists saw the Spemann organizer as the core element for the organization of the vertebrate embryonic axis and, instead, provides opportunities for the experimental control of morphogenesis starting with a population of embryonic pluripotent cells that will be instructed using those two morphogen gradients.
Collapse
Affiliation(s)
- Bernard Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Christine Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
24
|
Wu BT, Wen SH, Hwang SPL, Huang CJ, Kuan YS. Control of Wnt5b secretion by Wntless modulates chondrogenic cell proliferation through fine-tuning fgf3 expression. J Cell Sci 2015; 128:2328-39. [PMID: 25934698 DOI: 10.1242/jcs.167403] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
Wnts and Fgfs regulate various tissues development in vertebrates. However, how regional Wnt or Fgf activities are established and how they interact in any given developmental event is elusive. Here, we investigated the Wnt-mediated craniofacial cartilage development in zebrafish and found that fgf3 expression in the pharyngeal pouches is differentially reduced along the anteroposterior axis in wnt5b mutants and wntless (wls) morphants, but its expression is normal in wnt9a and wnt11 morphants. Introducing fgf3 mRNAs rescued the cartilage defects in Wnt5b- and Wls-deficient larvae. In wls morphants, endogenous Wls expression is not detectable but maternally deposited Wls is present in eggs, which might account for the lack of axis defects in wls morphants. Secretion of endogenous Wnt5b but not Wnt11 was affected in the pharyngeal tissue of Wls morphants, indicating that Wls is not involved in every Wnt secretion event. Furthermore, cell proliferation but not apoptosis in the developing jaw was affected in Wnt5b- and Wls-deficient embryos. Therefore, Wnt5b requires Wls for its secretion and regulates the proliferation of chondrogenic cells through fine-tuning the expression of fgf3 during jaw cartilage development.
Collapse
Affiliation(s)
- Bo-Tsung Wu
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Shih-Hsien Wen
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Sheng-Ping L Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chang-Jen Huang
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Yung-Shu Kuan
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan Center for System Biology, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
25
|
Larraguibel J, Weiss ARE, Pasula DJ, Dhaliwal RS, Kondra R, Van Raay TJ. Wnt ligand-dependent activation of the negative feedback regulator Nkd1. Mol Biol Cell 2015; 26:2375-84. [PMID: 25904337 PMCID: PMC4462952 DOI: 10.1091/mbc.e14-12-1648] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/16/2015] [Indexed: 02/02/2023] Open
Abstract
Nkd1, a negative feedback regulator of the Wnt pathway, localizes with Dvl2 to the putative Wnt signalosome, where it becomes activated by Wnt. Activated Nkd1 moves away from the membrane to become more cytosolic, where it interacts with β-catenin to prevent nuclear accumulation. Misregulation of Wnt signaling is at the root of many diseases, most notably colorectal cancer, and although we understand the activation of the pathway, we have a very poor understanding of the circumstances under which Wnt signaling turns itself off. There are numerous negative feedback regulators of Wnt signaling, but two stand out as constitutive and obligate Wnt-induced regulators: Axin2 and Nkd1. Whereas Axin2 behaves similarly to Axin in the destruction complex, Nkd1 is more enigmatic. Here we use zebrafish blastula cells that are responsive Wnt signaling to demonstrate that Nkd1 activity is specifically dependent on Wnt ligand activation of the receptor. Furthermore, our results support the hypothesis that Nkd1 is recruited to the Wnt signalosome with Dvl2, where it becomes activated to move into the cytoplasm to interact with β-catenin, inhibiting its nuclear accumulation. Comparison of these results with Nkd function in Drosophila generates a unified and conserved model for the role of this negative feedback regulator in the modulation of Wnt signaling.
Collapse
Affiliation(s)
- Jahdiel Larraguibel
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Alexander R E Weiss
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Daniel J Pasula
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Rasmeet S Dhaliwal
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Roman Kondra
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Terence J Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
26
|
Walker MP, Stopford CM, Cederlund M, Fang F, Jahn C, Rabinowitz AD, Goldfarb D, Graham DM, Yan F, Deal AM, Fedoriw Y, Richards KL, Davis IJ, Weidinger G, Damania B, Major MB. FOXP1 potentiates Wnt/β-catenin signaling in diffuse large B cell lymphoma. Sci Signal 2015; 8:ra12. [PMID: 25650440 DOI: 10.1126/scisignal.2005654] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transcription factor FOXP1 (forkhead box protein P1) is a master regulator of stem and progenitor cell biology. In diffuse large B cell lymphoma (DLBCL), copy number amplifications and chromosomal translocations result in overexpression of FOXP1. Increased abundance of FOXP1 in DLBCL is a predictor of poor prognosis and resistance to therapy. We developed a genome-wide, mass spectrometry-coupled, gain-of-function genetic screen, which revealed that FOXP1 potentiates β-catenin-dependent, Wnt-dependent gene expression. Gain- and loss-of-function studies in cell models and zebrafish confirmed that FOXP1 was a general and conserved enhancer of Wnt signaling. In a Wnt-dependent fashion, FOXP1 formed a complex with β-catenin, TCF7L2 (transcription factor 7-like 2), and the acetyltransferase CBP [CREB (adenosine 3',5'-monophosphate response element-binding protein)-binding protein], and this complex bound the promoters of Wnt target genes. FOXP1 promoted the acetylation of β-catenin by CBP, and acetylation was required for FOXP1-mediated potentiation of β-catenin-dependent transcription. In DLBCL, we found that FOXP1 promoted sensitivity to Wnt pathway inhibitors, and knockdown of FOXP1 or blocking β-catenin transcriptional activity slowed xenograft tumor growth. These data connect excessive FOXP1 with β-catenin-dependent signal transduction and provide a molecular rationale for Wnt-directed therapy in DLBCL.
Collapse
Affiliation(s)
- Matthew P Walker
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Charles M Stopford
- Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Maria Cederlund
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Fang Fang
- Carolina Center for Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Christopher Jahn
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Alex D Rabinowitz
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Dennis Goldfarb
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3175, USA
| | - David M Graham
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Feng Yan
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Allison M Deal
- UNC Lineberger Comprehensive Cancer Center Biostatistics Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Yuri Fedoriw
- Department of Pathology and Laboratory, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Kristy L Richards
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Ian J Davis
- Carolina Center for Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Blossom Damania
- Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. Division of Microbiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27516-7361, USA.
| |
Collapse
|
27
|
Filopodia-based Wnt transport during vertebrate tissue patterning. Nat Commun 2015; 6:5846. [PMID: 25556612 DOI: 10.1038/ncomms6846] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023] Open
Abstract
Paracrine Wnt/β-catenin signalling is important during developmental processes, tissue regeneration and stem cell regulation. Wnt proteins are morphogens, which form concentration gradients across responsive tissues. Little is known about the transport mechanism for these lipid-modified signalling proteins in vertebrates. Here we show that Wnt8a is transported on actin-based filopodia to contact responding cells and activate signalling during neural plate formation in zebrafish. Cdc42/N-Wasp regulates the formation of these Wnt-positive filopodia. Enhanced formation of filopodia increases the effective signalling range of Wnt by facilitating spreading. Consistently, reduction in filopodia leads to a restricted distribution of the ligand and a limited signalling range. Using a simulation, we provide evidence that such a short-range transport system for Wnt has a long-range signalling function. Indeed, we show that a filopodia-based transport system for Wnt8a controls anteroposterior patterning of the neural plate during vertebrate gastrulation.
Collapse
|
28
|
Green D, Whitener AE, Mohanty S, Lekven AC. Vertebrate nervous system posteriorization: Grading the function of Wnt signaling. Dev Dyn 2014; 244:507-12. [DOI: 10.1002/dvdy.24230] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 10/31/2014] [Accepted: 11/12/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- David Green
- Department of Biology; Texas A&M University; College Station Texas
| | - Amy E. Whitener
- Department of Biology; Texas A&M University; College Station Texas
| | - Saurav Mohanty
- Department of Biology; Texas A&M University; College Station Texas
| | - Arne C. Lekven
- Department of Biology; Texas A&M University; College Station Texas
| |
Collapse
|
29
|
Tissue-specific derepression of TCF/LEF controls the activity of the Wnt/β-catenin pathway. Nat Commun 2014; 5:5368. [PMID: 25371059 DOI: 10.1038/ncomms6368] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/24/2014] [Indexed: 11/09/2022] Open
Abstract
Upon stimulation by Wnt ligands, the canonical Wnt/β-catenin signalling pathway results in the stabilization of β-catenin and its translocation into the nucleus to form transcriptionally active complexes with sequence-specific DNA-binding T-cell factor/lymphoid enhancer factor (TCF/LEF) family proteins. In the absence of nuclear β-catenin, TCF proteins act as transcriptional repressors by binding to Groucho/Transducin-Like Enhancer of split (TLE) proteins that function as co-repressors by interacting with histone deacetylases whose activity leads to the generation of transcriptionally silent chromatin. Here we show that the transcription factor Ladybird homeobox 2 (Lbx2) positively controls the Wnt/β-catenin signalling pathway in the posterior lateral and ventral mesoderm of the zebrafish embryo at the gastrula stage, by directly interfering with the binding of Groucho/TLE to TCF, thereby preventing formation of transcription repressor complexes. These findings reveal a novel level of regulation of the canonical Wnt/β-catenin signalling pathway occurring in the nucleus and involving tissue-specific derepression of TCF by Lbx2.
Collapse
|
30
|
Feng L, Jiang H, Wu P, Marlow FL. Negative feedback regulation of Wnt signaling via N-linked fucosylation in zebrafish. Dev Biol 2014; 395:268-86. [PMID: 25238963 DOI: 10.1016/j.ydbio.2014.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 07/25/2014] [Accepted: 09/09/2014] [Indexed: 01/05/2023]
Abstract
L-fucose, a monosaccharide widely distributed in eukaryotes and certain bacteria, is a determinant of many functional glycans that play central roles in numerous biological processes. The molecular mechanism, however, by which fucosylation mediates these processes remains largely elusive. To study how changes in fucosylation impact embryonic development, we up-regulated N-linked fucosylation via over-expression of a key GDP-Fucose transporter, Slc35c1, in zebrafish. We show that Slc35c1 overexpression causes elevated N-linked fucosylation and disrupts embryonic patterning in a transporter activity dependent manner. We demonstrate that patterning defects associated with enhanced N-linked fucosylation are due to diminished canonical Wnt signaling. Chimeric analyses demonstrate that elevated Slc35c1 expression in receiving cells decreases the signaling range of Wnt8a during zebrafish embryogenesis. Moreover, we provide biochemical evidence that this decrease is associated with reduced Wnt8 ligand and elevated Lrp6 coreceptor, which we show are both substrates for N-linked fucosylation in zebrafish embryos. Strikingly, slc35c1 expression is regulated by canonical Wnt signaling. These results suggest that Wnt limits its own signaling activity in part via up-regulation of a transporter, slc35c1 that promotes terminal fucosylation and thereby limits Wnt activity.
Collapse
Affiliation(s)
- Lei Feng
- Department of Biochemistry, Albert Einstein College of Medicine Yeshiva University, Bronx, NY 10461, USA
| | - Hao Jiang
- Department of Biochemistry, Albert Einstein College of Medicine Yeshiva University, Bronx, NY 10461, USA
| | - Peng Wu
- Department of Biochemistry, Albert Einstein College of Medicine Yeshiva University, Bronx, NY 10461, USA.
| | - Florence L Marlow
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine Yeshiva University, Bronx, NY 10461, USA
| |
Collapse
|
31
|
Shimizu N, Ishitani S, Sato A, Shibuya H, Ishitani T. Hipk2 and PP1c Cooperate to Maintain Dvl Protein Levels Required for Wnt Signal Transduction. Cell Rep 2014; 8:1391-404. [DOI: 10.1016/j.celrep.2014.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 06/04/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022] Open
|
32
|
Hagemann AIH, Kurz J, Kauffeld S, Chen Q, Reeves PM, Weber S, Schindler S, Davidson G, Kirchhausen T, Scholpp S. In vivo analysis of formation and endocytosis of the Wnt/β-catenin signaling complex in zebrafish embryos. J Cell Sci 2014; 127:3970-82. [PMID: 25074807 PMCID: PMC4163645 DOI: 10.1242/jcs.148767] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
After activation by Wnt/β-Catenin ligands, a multi-protein complex assembles at the clustering membrane-bound receptors and intracellular signal transducers into the so-called Lrp6-signalosome. However, the mechanism of signalosome formation and dissolution is yet not clear. Our imaging studies of live zebrafish embryos show that the signalosome is a highly dynamic structure. It is continuously assembled by Dvl2-mediated recruitment of the transducer complex to the activated receptors and partially disassembled by endocytosis. We find that, after internalization, the ligand-receptor complex and the transducer complex take separate routes. The Wnt–Fz–Lrp6 complex follows a Rab-positive endocytic path. However, when still bound to the transducer complex, Dvl2 forms intracellular aggregates. We show that this endocytic process is not only essential for ligand-receptor internalization but also for signaling. The μ2-subunit of the endocytic Clathrin adaptor Ap2 interacts with Dvl2 to maintain its stability during endocytosis. Blockage of Ap2μ2 function leads to Dvl2 degradation, inhibiton of signalosome formation at the plasma membrane and, consequently, reduction of signaling. We conclude that Ap2μ2-mediated endocytosis is important to maintain Wnt/β-catenin signaling in vertebrates.
Collapse
Affiliation(s)
- Anja I H Hagemann
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Jennifer Kurz
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Silke Kauffeld
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Qing Chen
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Patrick M Reeves
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, 02115 MA, USA
| | - Sabrina Weber
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Simone Schindler
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Gary Davidson
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Tomas Kirchhausen
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, 02115 MA, USA
| | - Steffen Scholpp
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| |
Collapse
|
33
|
Rong X, Chen C, Zhou P, Zhou Y, Li Y, Lu L, Liu Y, Zhou J, Duan C. R-spondin 3 regulates dorsoventral and anteroposterior patterning by antagonizing Wnt/β-catenin signaling in zebrafish embryos. PLoS One 2014; 9:e99514. [PMID: 24918770 PMCID: PMC4053527 DOI: 10.1371/journal.pone.0099514] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
The Wnt/β-catenin or canonical Wnt signaling pathway plays fundamental roles in early development and in maintaining adult tissue homeostasis. R-spondin 3 (Rspo3) is a secreted protein that has been implicated in activating the Wnt/β-catenin signaling in amphibians and mammals. Here we report that zebrafish Rspo3 plays a negative role in regulating the zygotic Wnt/β-catenin signaling. Zebrafish Rspo3 has a unique domain structure. It contains a third furin-like (FU3) domain. This FU3 is present in other four ray-finned fish species studied but not in elephant shark. In zebrafish, rspo3 mRNA is maternally deposited and has a ubiquitous expression in early embryonic stages. After 12 hpf, its expression becomes tissue-specific. Forced expression of rspo3 promotes dorsoanterior patterning and increases the expression of dorsal and anterior marker genes. Knockdown of rspo3 increases ventral-posterior development and stimulates ventral and posterior marker genes expression. Forced expression of rspo3 abolishes exogenous Wnt3a action and reduces the endogenous Wnt signaling activity. Knockdown of rspo3 results in increased Wnt/β-catenin signaling activity. Further analyses indicate that Rspo3 does not promote maternal Wnt signaling. Human RSPO3 has similar action when tested in zebrafish embryos. These results suggest that Rspo3 regulates dorsoventral and anteroposterior patterning by negatively regulating the zygotic Wnt/β-catenin signaling in zebrafish embryos.
Collapse
Affiliation(s)
- Xiaozhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Chen Chen
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Pin Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yumei Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
- * E-mail: (CD); (JZ)
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (CD); (JZ)
| |
Collapse
|
34
|
Chen JB, Gao HW, Zhang YL, Zhang Y, Zhou XF, Li CQ, Gao HP. Developmental toxicity of diclofenac and elucidation of gene regulation in zebrafish (Danio rerio). Sci Rep 2014; 4:4841. [PMID: 24788080 PMCID: PMC4007093 DOI: 10.1038/srep04841] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 04/08/2014] [Indexed: 01/08/2023] Open
Abstract
Environmental pollution by emerging contaminants, e.g. pharmaceuticals, has become a matter of widespread concern in recent years. We investigated the membrane transport of diclofenac and its toxic effects on gene expression and the development of zebrafish embryos. The association of diclofenac with the embryos conformed to the general partition model at low concentration, the partition coefficient being 0.0033 ml per embryo. At high concentration, the interaction fitted the Freundlich model. Most of the diclofenac remained in the extracellular aqueous solution with less than 5% interacting with the embryo, about half of which was adsorbed on the membranes while the rest entered the cytoplasm. Concentrations of diclofenac over 10.13 μM were lethal to all the embryos, while 3.78 μM diclofenac was teratogenic. The development abnormalities at 4 day post treatment (dpt) include shorter body length, smaller eye, pericardial and body edema, lack of liver, intestine and circulation, muscle degeneration, and abnormal pigmentation. The portion of the diclofenac transferred into the embryo altered the expression of certain genes, e.g. down-regulation of Wnt3a and Gata4 and up-regulation of Wnt8a. The alteration of expression of such genes or the regulation of downstream genes could cause defects in the cardiovascular and nervous systems.
Collapse
Affiliation(s)
- Jia-Bin Chen
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Hong-Wen Gao
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Ya-Lei Zhang
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yong Zhang
- Hunter Biotechnology, Inc., Hangzhou, 311231, China
| | - Xue-Fei Zhou
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Chun-Qi Li
- Hunter Biotechnology, Inc., Hangzhou, 311231, China
| | - Hai-Ping Gao
- State Key Laboratory of Pollution Control and Resources Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| |
Collapse
|
35
|
Bajard L, Morelli LG, Ares S, Pécréaux J, Jülicher F, Oates AC. Wnt-regulated dynamics of positional information in zebrafish somitogenesis. Development 2014; 141:1381-91. [PMID: 24595291 PMCID: PMC3943186 DOI: 10.1242/dev.093435] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
How signaling gradients supply positional information in a field of moving cells is an unsolved question in patterning and morphogenesis. Here, we ask how a Wnt signaling gradient regulates the dynamics of a wavefront of cellular change in a flow of cells during somitogenesis. Using time-controlled perturbations of Wnt signaling in the zebrafish embryo, we changed segment length without altering the rate of somite formation or embryonic elongation. This result implies specific Wnt regulation of the wavefront velocity. The observed Wnt signaling gradient dynamics and timing of downstream events support a model for wavefront regulation in which cell flow plays a dominant role in transporting positional information.
Collapse
Affiliation(s)
- Lola Bajard
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Mwafi N, Beretta CA, Paolini A, Carl M. Divergent Wnt8a gene expression in teleosts. PLoS One 2014; 9:e85303. [PMID: 24465531 PMCID: PMC3896364 DOI: 10.1371/journal.pone.0085303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/26/2013] [Indexed: 12/02/2022] Open
Abstract
The analysis of genes in evolutionarily distant but morphologically similar species is of major importance to unravel the changes in genomes over millions of years, which led to gene silencing and functional diversification. We report the analysis of Wnt8a gene expression in the medakafish and provide a detailed comparison to other vertebrates. In all teleosts analyzed there are two paralogous Wnt8a copies. These show largely overlapping expression in the early developing zebrafish embryo, an evolutionarily distant relative of medaka. In contrast to zebrafish, we find that both maternal and zygotic expression of particularly one Wnt8a paralog has diverged in medaka. While Wnt8a1 expression is mostly conserved at early embryonic stages, the expression of Wnt8a2 differs markedly. In addition, both genes are distinctly expressed during organogenesis unlike the zebrafish homologs, which may hint at the emergence of functional diversification of Wnt8a ligands during evolution.
Collapse
Affiliation(s)
- Nesrin Mwafi
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Mannheim, Germany
| | - Carlo A. Beretta
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Mannheim, Germany
| | - Alessio Paolini
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Mannheim, Germany
| | - Matthias Carl
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Mannheim, Germany
- * E-mail:
| |
Collapse
|
37
|
Kapp LD, Abrams EW, Marlow FL, Mullins MC. The integrator complex subunit 6 (Ints6) confines the dorsal organizer in vertebrate embryogenesis. PLoS Genet 2013; 9:e1003822. [PMID: 24204286 PMCID: PMC3814294 DOI: 10.1371/journal.pgen.1003822] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 08/08/2013] [Indexed: 11/19/2022] Open
Abstract
Dorsoventral patterning of the embryonic axis relies upon the mutual antagonism of competing signaling pathways to establish a balance between ventralizing BMP signaling and dorsal cell fate specification mediated by the organizer. In zebrafish, the initial embryo-wide domain of BMP signaling is refined into a morphogenetic gradient following activation dorsally of a maternal Wnt pathway. The accumulation of β-catenin in nuclei on the dorsal side of the embryo then leads to repression of BMP signaling dorsally and the induction of dorsal cell fates mediated by Nodal and FGF signaling. A separate Wnt pathway operates zygotically via Wnt8a to limit dorsal cell fate specification and maintain the expression of ventralizing genes in ventrolateral domains. We have isolated a recessive dorsalizing maternal-effect mutation disrupting the gene encoding Integrator Complex Subunit 6 (Ints6). Due to widespread de-repression of dorsal organizer genes, embryos from mutant mothers fail to maintain expression of BMP ligands, fail to fully express vox and ved, two mediators of Wnt8a, display delayed cell movements during gastrulation, and severe dorsalization. Consistent with radial dorsalization, affected embryos display multiple independent axial domains along with ectopic dorsal forerunner cells. Limiting Nodal signaling or restoring BMP signaling restores wild-type patterning to affected embryos. Our results are consistent with a novel role for Ints6 in restricting the vertebrate organizer to a dorsal domain in embryonic patterning.
Collapse
Affiliation(s)
- Lee D. Kapp
- Perelman School of Medicine at the University of Pennsylvania, Department of Cell and Developmental Biology, Philadelphia, Pennsylvania, United States of America
| | - Elliott W. Abrams
- Perelman School of Medicine at the University of Pennsylvania, Department of Cell and Developmental Biology, Philadelphia, Pennsylvania, United States of America
| | - Florence L. Marlow
- Perelman School of Medicine at the University of Pennsylvania, Department of Cell and Developmental Biology, Philadelphia, Pennsylvania, United States of America
| | - Mary C. Mullins
- Perelman School of Medicine at the University of Pennsylvania, Department of Cell and Developmental Biology, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
38
|
Warga RM, Mueller RL, Ho RK, Kane DA. Zebrafish Tbx16 regulates intermediate mesoderm cell fate by attenuating Fgf activity. Dev Biol 2013; 383:75-89. [PMID: 24008197 DOI: 10.1016/j.ydbio.2013.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/04/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
Abstract
Progenitors of the zebrafish pronephros, red blood and trunk endothelium all originate from the ventral mesoderm and often share lineage with one another, suggesting that their initial patterning is linked. Previous studies have shown that spadetail (spt) mutant embryos, defective in tbx16 gene function, fail to produce red blood cells, but retain the normal number of endothelial and pronephric cells. We report here that spt mutants are deficient in all the types of early blood, have fewer endothelial cells as well as far more pronephric cells compared to wildtype. In vivo cell tracing experiments reveal that blood and endothelium originate in spt mutants almost exclusive from the dorsal mesoderm whereas, pronephros and tail originate from both dorsal and ventral mesoderm. Together these findings suggest possible defects in posterior patterning. In accord with this, gene expression analysis shows that mesodermal derivatives within the trunk and tail of spt mutants have acquired more posterior identity. Secreted signaling molecules belonging to the Fgf, Wnt and Bmp families have been implicated as patterning factors of the posterior mesoderm. Further investigation demonstrates that Fgf and Wnt signaling are elevated throughout the nonaxial region of the spt gastrula. By manipulating Fgf signaling we show that Fgfs both promote pronephric fate and repress blood and endothelial fate. We conclude that Tbx16 plays an important role in regulating the balance of intermediate mesoderm fates by attenuating Fgf activity.
Collapse
Affiliation(s)
- Rachel M Warga
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA; Department of Organismal Biology and Anatomy, University of Chicago, 1027 East, 57th Street, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
39
|
Angonin D, Van Raay TJ. Nkd1 functions as a passive antagonist of Wnt signaling. PLoS One 2013; 8:e74666. [PMID: 24009776 PMCID: PMC3756965 DOI: 10.1371/journal.pone.0074666] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/05/2013] [Indexed: 12/22/2022] Open
Abstract
Wnt signaling is involved in many aspects of development and in the homeostasis of stem cells. Its importance is underscored by the fact that misregulation of Wnt signaling has been implicated in numerous diseases, especially colorectal cancer. However, how Wnt signaling regulates itself is not well understood. There are several Wnt negative feedback regulators, which are active antagonists of Wnt signaling, but one feedback regulator, Nkd1, has reduced activity compared to other antagonists, yet is still a negative feedback regulator. Here we describe our efforts to understand the role of Nkd1 using Wnt signaling compromised zebrafish mutant lines. In several of these lines, Nkd1 function was not any more active than it was in wild type embryos. However, we found that Nkd1’s ability to antagonize canonical Wnt/β-catenin signaling was enhanced in the Wnt/Planar Cell Polarity mutants silberblick (slb/wnt11) and trilobite (tri/vangl2). While slb and tri mutants do not display alterations in canonical Wnt signaling, we found that they are hypersensitive to it. Overexpression of the canonical Wnt/β-catenin ligand Wnt8a in slb or tri mutants resulted in dorsalized embryos, with tri mutants being much more sensitive to Wnt8a than slb mutants. Furthermore, the hyperdorsalization caused by Wnt8a in tri could be rescued by Nkd1. These results suggest that Nkd1 functions as a passive antagonist of Wnt signaling, functioning only when homeostatic levels of Wnt signaling have been breached or when Wnt signaling becomes destabilized.
Collapse
Affiliation(s)
- Diane Angonin
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Terence J. Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
40
|
Wolf A, Ryu S. Specification of posterior hypothalamic neurons requires coordinated activities of Fezf2, Otp, Sim1a and Foxb1.2. Development 2013; 140:1762-73. [PMID: 23533176 DOI: 10.1242/dev.085357] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The hypothalamus is a key integrative center in the brain that consists of diverse cell types required for a variety of functions including homeostasis, reproduction, stress response, social and cognitive behavior. Despite our knowledge of several transcription factors crucial for hypothalamic development, it is not known how the wide diversity of neuron types in the hypothalamus is produced. In particular, almost nothing is known about the mechanisms that specify neurons in the posteriormost part of the hypothalamus, the mammillary area. Here, we investigated the specification of two distinct neuron types in the mammillary area that produce the hypothalamic hormones Vasoactive intestinal peptide (Vip) and Urotensin 1 (Uts1). We show that Vip- and Uts1-positive neurons develop in distinct domains in the mammillary area defined by the differential expression of the transcription factors Fezf2, Otp, Sim1a and Foxb1.2. Coordinated activities of these factors are crucial for the establishment of the mammillary area subdomains and the specification of Vip- and Uts1-positive neurons. In addition, Fezf2 is important for early development of the posterior hypothalamus. Thus, our study provides the first molecular anatomical map of the posterior hypothalamus in zebrafish and identifies, for the first time, molecular requirements underlying the specification of distinct posterior hypothalamic neuron types.
Collapse
Affiliation(s)
- Andrea Wolf
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | |
Collapse
|
41
|
O'Neill K, Thorpe C. BMP signaling and spadetail regulate exit of muscle precursors from the zebrafish tailbud. Dev Biol 2013; 375:117-27. [PMID: 23246591 DOI: 10.1016/j.ydbio.2012.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 11/26/2012] [Accepted: 12/04/2012] [Indexed: 11/19/2022]
Abstract
The tailbud is a population of stem cells in the posterior embryonic tail. During zebrafish development, these stem cells give rise to the main structures of the embryo's posterior body, including the tail somites. Progenitor cells reside in the tailbud for variable amounts of time before they exit and begin to differentiate. There must be a careful balance between cells that leave the tailbud and cells that are held back in order to give rise to later somites. However, this meticulous process is not well understood. A gene that has shed some light on this area is the t-box transcription factor spadetail (spt). When spt is mutated, embryos develop an enlarged tailbud and are only able to form roughly half of their somites. This phenotype is due to the fact that some of the somitic precursors are not able to leave the tailbud or differentiate. Another factor involved in tail morphogenesis is the Bone Morphogenetic Protein (BMP) pathway. BMPs are important for many processes during early development, including cell migration. Chordino (chd) is a secreted protein that inhibits BMP signaling. BMPs are upregulated in chd mutants, however, these mutants are able to form organized somites. In embryos where chd and spt are mutated, somites are completely absent. These double mutants also develop a large tailbud due to the accumulation of progenitor cells that are never able to leave or differentiate. To study the dynamics of cells in the tailbud and their role in somite formation, we have analyzed the genetic factors and pathway interactions involved, conducted transplant experiments to look at behavior of mutant cells in different genetic backgrounds, and used time lapse microscopy to characterize cell movements and behavior in wild type and mutant tailbuds. These data suggest that spt expression and BMP inhibition are both required for somitic precursors to exit the tailbud. They also elucidate that chd;spt tailbud mesodermal progenitor cells (MPC) behave autonomously and their dynamics within the tailbud are drastically different than WT MPCs.
Collapse
Affiliation(s)
- Katelyn O'Neill
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | | |
Collapse
|
42
|
Mori S, Moriyama Y, Yoshikawa K, Furukawa T, Kuroda H. β-Adrenergic signaling promotes posteriorization in Xenopus early development. Dev Growth Differ 2013; 55:350-8. [PMID: 23452088 DOI: 10.1111/dgd.12046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/05/2013] [Accepted: 01/15/2013] [Indexed: 11/29/2022]
Abstract
Adrenaline (also known as Epinephrine) is a hormone, which works as major regulator of various biological events such stages of vertebrate, the role of adrenaline for early embryogenesis has been as heart rate, blood vessel and air passage diameters, and metabolic shifts. Although its specific receptors are expressing at the early developmental stage those functions are poorly understood. Here, we show that loss-of-functional effects of adrenergic receptor β-2 (Adrβ2), which was known as the major receptor for adrenaline and highly expressed in embryonic stages, led posterior defects at the tadpole stage of Xenopus embryos, while embryos injected with Adrβ2 mRNA or treated with adrenaline hormone adversely lost anterior structures. This posteriorization effect by adrenaline hormone was dose-dependently increased but effectively rescued by microinjection of antisense morpholino oligomer for Adrβ2 (Adrβ2-MO). Combination of adrenaline treatments and microinjection of Adrβ2 mRNA maximized efficiency in its posteriorizing activity. Interestingly, both gain- and loss-of-functional treatment for β-adrenergic signaling could not influence anterior neural fate induced by overexpression of Chordin mRNA in presumptive ectodermal region, meaning that it worked via mesoderm. Taken together with these results, we conclude that adrenaline is a novel regulator of anteroposterior axis formation in vertebrates.
Collapse
Affiliation(s)
- Shoko Mori
- Graduate School of Science and Technology, Shizuoka University, 836 Ohya Suruga-ku, Shizuoka, 422-8529, Japan
| | | | | | | | | |
Collapse
|
43
|
Hofmeister W, Key B. Frizzled-3a and Wnt-8b genetically interact during forebrain commissural formation in embryonic zebrafish. Brain Res 2013; 1506:25-34. [PMID: 23438515 DOI: 10.1016/j.brainres.2013.02.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 02/01/2013] [Accepted: 02/15/2013] [Indexed: 12/31/2022]
Abstract
The commissural plate forms the rostral surface of the embryonic vertebrate forebrain and provides a cellular substrate for forebrain commissural axons. We have previously reported that the Wnt receptor frizzled-3a (fzd3a) restricts the expression of the chemorepulsive guidance ligand slit2 to a discrete domain of neuroepithelial cells in the commissural plate of embryonic zebrafish. Loss of Fzd3a function perturbed slit2 expression and disrupted the formation of glial bridges which guide the formation of forebrain commissures. We now show that Wnt8b is also necessary for anterior commissural formation as well as for patterning of slit2 expression at the midline. Knock down of Wnt8b produced the same phenotype as loss of Fzd3a which suggested that these genes were acting together to regulate axon guidance. Simultaneous sub-threshold knock down of both Fzd3a and Wnt8b led to a greater than additive increase in the penetrance of the mutant phenotype which indicated that these two genes were indeed interacting. We have shown here that Fzd3a/Wnt8b signaling is essential for normal patterning of the commissural plate and that loss-of-function in either receptor or ligand causes Slit2-dependent defects in glial bridge morphology which indirectly attenuated axon midline crossing in the embryonic vertebrate forebrain.
Collapse
Affiliation(s)
- Wolfgang Hofmeister
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | | |
Collapse
|
44
|
Liu JX, Zhang D, Xie X, Ouyang G, Liu X, Sun Y, Xiao W. Eaf1 and Eaf2 negatively regulate canonical Wnt/β-catenin signaling. Development 2013; 140:1067-78. [PMID: 23364330 DOI: 10.1242/dev.086157] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Eaf factors play a crucial role in tumor suppression and embryogenesis. To investigate the potential mechanism of Eaf activity, we performed loss- and gain-of-function assays in zebrafish using morpholino and mRNA injections, respectively. We found that eaf1 and eaf2 inhibit Wnt/β-catenin signaling, thereby modulating mesodermal and neural patterning in the embryo. Moreover, ectopic expression of eaf1 and eaf2 in embryos and cultured cells blocked β-catenin reporter activity. By immunoprecipitation, we also observed that Eaf1 and Eaf2 bound to the Armadillo repeat region and C-terminus of β-catenin, as well as to other β-catenin transcription complex proteins, such as c-Jun, Tcf and Axin, suggesting the formation of a novel complex. In addition, the N-terminus of Eaf1 and Eaf2 bound to β-catenin and exhibited dominant-negative activity, whereas the C-terminus appeared to either harbor a suppression domain or to recruit a repressor. Both the N- and C-terminus must be intact for Eaf1 and Eaf2 suppressive activity. Lastly, we demonstrate a conservation of biological activities for Eaf family proteins across species. In summary, our evidence points to a novel role for Eaf1 and Eaf2 in inhibiting canonical Wnt/β-catenin signaling, which might form the mechanistic basis for Eaf1 and Eaf2 tumor suppressor activity.
Collapse
Affiliation(s)
- Jing-Xia Liu
- Key Laboratory of Biodiversity and Conservation of Aquatic Organisms, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, PR China
| | | | | | | | | | | | | |
Collapse
|
45
|
Nakamura T, Saito D, Kawasumi A, Shinohara K, Asai Y, Takaoka K, Dong F, Takamatsu A, Belo JA, Mochizuki A, Hamada H. Fluid flow and interlinked feedback loops establish left–right asymmetric decay of Cerl2 mRNA. Nat Commun 2012; 3:1322. [DOI: 10.1038/ncomms2319] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 11/22/2012] [Indexed: 11/09/2022] Open
|
46
|
Feng Q, Zou X, Lu L, Li Y, Liu Y, Zhou J, Duan C. The stress-response gene redd1 regulates dorsoventral patterning by antagonizing Wnt/β-catenin activity in zebrafish. PLoS One 2012; 7:e52674. [PMID: 23300740 PMCID: PMC3530439 DOI: 10.1371/journal.pone.0052674] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 11/19/2012] [Indexed: 11/25/2022] Open
Abstract
REDD1/redd1 is a stress-response gene that is induced under various stressful conditions such as hypoxia, DNA damage, and energy stress. The increased REDD1 inhibits mTOR signaling and cell growth. Here we report an unexpected role of Redd1 in regulating dorsoventral patterning in zebrafish embryos and the underlying mechanisms. Zebrafish redd1 mRNA is maternally deposited. Although it is ubiquitously detected in many adult tissues, its expression is highly tissue-specific and dynamic during early development. Hypoxia and heat shock strongly induce redd1 expression in zebrafish embryos. Knockdown of Redd1 using two independent morpholinos results in dorsalized embryos and this effect can be rescued by injecting redd1 mRNA. Forced expression of Redd1 ventralizes embryos. Co-expression of Redd1 with Wnt3a or a constitutively active form of β-catenin suggests that Redd1 alters dorsoventral patterning by antagonizing the Wnt/β-catenin signaling pathway. These findings have unraveled a novel role of Redd1 in early development by antagonizing Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Qiang Feng
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Xia Zou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
- * E-mail: (JZ); (CD)
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (JZ); (CD)
| |
Collapse
|
47
|
Carlin D, Sepich D, Grover VK, Cooper MK, Solnica-Krezel L, Inbal A. Six3 cooperates with Hedgehog signaling to specify ventral telencephalon by promoting early expression of Foxg1a and repressing Wnt signaling. Development 2012; 139:2614-24. [PMID: 22736245 PMCID: PMC3383232 DOI: 10.1242/dev.076018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2012] [Indexed: 01/18/2023]
Abstract
Six3 exerts multiple functions in the development of anterior neural tissue of vertebrate embryos. Whereas complete loss of Six3 function in the mouse results in failure of forebrain formation, its hypomorphic mutations in human and mouse can promote holoprosencephaly (HPE), a forebrain malformation that results, at least in part, from abnormal telencephalon development. However, the roles of Six3 in telencephalon patterning and differentiation are not well understood. To address the role of Six3 in telencephalon development, we analyzed zebrafish embryos deficient in two out of three Six3-related genes, six3b and six7, representing a partial loss of Six3 function. We found that telencephalon forms in six3b;six7-deficient embryos; however, ventral telencephalic domains are smaller and dorsal domains are larger. Decreased cell proliferation or excess apoptosis cannot account for the ventral deficiency. Instead, six3b and six7 are required during early segmentation for specification of ventral progenitors, similar to the role of Hedgehog (Hh) signaling in telencephalon development. Unlike in mice, we observe that Hh signaling is not disrupted in embryos with reduced Six3 function. Furthermore, six3b overexpression is sufficient to compensate for loss of Hh signaling in isl1- but not nkx2.1b-positive cells, suggesting a novel Hh-independent role for Six3 in telencephalon patterning. We further find that Six3 promotes ventral telencephalic fates through transient regulation of foxg1a expression and repression of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Dan Carlin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Diane Sepich
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Vandana K. Grover
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael K. Cooper
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lilianna Solnica-Krezel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Adi Inbal
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Medical Neurobiology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
48
|
Narayanan A, Lekven AC. Biphasic wnt8a expression is achieved through interactions of multiple regulatory inputs. Dev Dyn 2012; 241:1062-75. [PMID: 22473868 DOI: 10.1002/dvdy.23787] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2012] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Vertebrate axis development depends upon wnt8a transcription in a dynamic pool of mesoderm progenitors at the posterior pole of the gastrulating embryo. The transcriptional mechanisms controlling wnt8a expression are not understood, but previous studies identified two phases of wnt8a expression in zebrafish: Nodal-dependent activation during early gastrulation (phase I) and No tail (Ntl)-dependent regulation from mid gastrula stages (phase II). RESULTS We identified two upstream cis-regulatory regions, proximal and distal, each of which possesses a promoter. The proximal regulatory region contains a margin-specific enhancer that is required for both the Nodal and Ntl responses. Phase I expression requires Nodal activation of the margin enhancer in combination with the transcription factor Zbtb4 and the distal regulatory region. Phase II expression requires Ntl regulation of the margin enhancer in the context of the proximal regulatory region. An additional mechanism is required to ensure the transition from phase I to phase II regulation. Analysis of stickleback wnt8a suggests this mechanism of regulation may be conserved. CONCLUSIONS The seemingly simple wnt8a expression pattern reflects complex interactions of multiple regulatory inputs.
Collapse
Affiliation(s)
- Anand Narayanan
- Department of Biology, Texas A&M University, College Station, Texas 77843-3258, USA
| | | |
Collapse
|
49
|
Abstract
Vertebrate development begins with precise molecular, cellular, and morphogenetic controls to establish the basic body plan of the embryo. In zebrafish, these tightly regulated processes begin during oogenesis and proceed through gastrulation to establish and pattern the axes of the embryo. During oogenesis a maternal factor is localized to the vegetal pole of the oocyte that is a determinant of dorsal tissues. Following fertilization this vegetally localized dorsal determinant is asymmetrically translocated in the egg and initiates formation of the dorsoventral axis. Dorsoventral axis formation and patterning is then mediated by maternal and zygotic factors acting through Wnt, BMP (bone morphogenetic protein), Nodal, and FGF (fibroblast growth factor) signaling pathways, each of which is required to establish and/or pattern the dorsoventral axis. This review addresses recent advances in our understanding of the molecular factors and mechanisms that establish and pattern the dorsoventral axis of the zebrafish embryo, including establishment of the animal-vegetal axis as it relates to formation of the dorsoventral axis.
Collapse
Affiliation(s)
- Yvette G Langdon
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
50
|
Identification and mechanism of regulation of the zebrafish dorsal determinant. Proc Natl Acad Sci U S A 2011; 108:15876-80. [PMID: 21911385 DOI: 10.1073/pnas.1106801108] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In vertebrates, the animal-vegetal axis is determined during oogenesis and at ovulation, the egg is radially symmetric. In anamniotes, following fertilization, a microtubule-dependent movement leads to the displacement of maternal dorsal determinants from the vegetal pole to the future dorsal side of the embryo, providing the initial breaking of radial symmetry [Weaver C, Kimelman D (2004) Development 131:3491-3499]. These dorsal determinants induce β-catenin nuclear translocation in dorsal cells of the blastula. Previous work in amphibians has shown that secreted Wnt11/5a complexes, regulated by the Wnt antagonist Dkk-1, are required for the initiation of embryonic axis formation [Cha et al. (2009) Curr Biol 29:1573-1580]. In the current study, we determined that the vegetal maternal dorsal determinant in fish is not the Wnt11/5a complex but the canonical Wnt, Wnt8a. Translation of this mRNA and secretion of the Wnt8a protein result in a dorsal-to-ventral gradient of Wnt stimulation, extending across the entire embryo. This gradient is counterbalanced by two Wnt inhibitors, Sfrp1a and Frzb. These proteins are essential to restrict the activation of the canonical Wnt pathway to the dorsal marginal blastomeres by defining the domain where the Wnt8a activity gradient is above the threshold value necessary for triggering the canonical β-catenin pathway. In summary, this study establishes that the zebrafish maternal dorsal determinant, Wnt8a, is required to localize the primary dorsal center, and that the extent of this domain is defined by the activity of two maternally provided Wnt antagonists, Sfrp1a and Frzb.
Collapse
|