1
|
Dicipulo R, Selland LG, Carpenter RG, Waskiewicz AJ. Functional role for Taz during hindbrain ventricle morphogenesis. PLoS One 2025; 20:e0313262. [PMID: 40080483 PMCID: PMC11906067 DOI: 10.1371/journal.pone.0313262] [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: 12/13/2023] [Accepted: 10/21/2024] [Indexed: 03/15/2025] Open
Abstract
The brain ventricle system, composed of the ventricular cavities and the cerebral spinal fluid within, performs critical functions including circulation of nutrients, removal of wastes, and cushioning of neural tissues. Development of the hindbrain ventricle requires a series of factors that coordinate its initial formation and subsequent inflation. Previous work has demonstrated that the transcriptional co-activator Taz (also known as WW domain-containing transcription regulator protein 1, Wwtr1), a component of Hippo signalling, is active at hindbrain rhombomere boundaries where it is regulated by mechanotransduction and promotes proliferation. Here, we demonstrate that Taz is also a critical regulator of hindbrain ventricle development. Zebrafish embryos that lack Taz protein fail to undergo initial midline separation of the hindbrain ventricle. Furthermore, the ventricle phenotype is a result of disorganized cytoskeletal F-actin and apicobasal polarity components. In addition, we have demonstrated that the hindbrain rhombomere boundaries are a location of active Wnt-Hippo crosstalk. Through our work, we propose a model where Taz protein is stabilized at rhombomere boundaries and promotes proper cell polarity necessary for formation of the brain ventricle.
Collapse
Affiliation(s)
- Renée Dicipulo
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Lyndsay G. Selland
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Rowan G. Carpenter
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J. Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Women & Children’s Health Research Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
2
|
Maekawa M, Saito S, Isobe D, Takemoto K, Miura Y, Dobashi Y, Yamasu K. The Oct4-related PouV gene, pou5f3, mediates isthmus development in zebrafish by directly and dynamically regulating pax2a. Cells Dev 2024; 179:203933. [PMID: 38908828 DOI: 10.1016/j.cdev.2024.203933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Using a transgenic zebrafish line harboring a heat-inducible dominant-interference pou5f3 gene (en-pou5f3), we reported that this PouV gene is involved in isthmus development at the midbrain-hindbrain boundary (MHB), which patterns the midbrain and cerebellum. Importantly, the functions of pou5f3 reportedly differ before and after the end of gastrulation. In the present study, we examined in detail the effects of en-pou5f3 induction on isthmus development during embryogenesis. When en-pou5f3 was induced around the end of gastrulation (bud stage), the isthmus was abrogated or deformed by the end of somitogenesis (24 hours post-fertilization). At this stage, the expression of MHB markers -- such as pax2a, fgf8a, wnt1, and gbx2 -- was absent in embryos lacking the isthmus structure, whereas it was present, although severely distorted, in embryos with a deformed isthmus. We further found that, after en-pou5f3 induction at late gastrulation, pax2a, fgf8a, and wnt1 were immediately and irreversibly downregulated, whereas the expression of en2a and gbx2 was reduced only weakly and slowly. Induction of en-pou5f3 at early somite stages also immediately downregulated MHB genes, particularly pax2a, but their expression was restored later. Overall, the data suggested that pou5f3 directly upregulates at least pax2a and possibly fgf8a and wnt1, which function in parallel in establishing the MHB, and that the role of pou5f3 dynamically changes around the end of gastrulation. We next examined the transcriptional regulation of pax2a using both in vitro and in vivo reporter analyses; the results showed that two upstream 1.0-kb regions with sequences conserved among vertebrates specifically drove transcription at the MHB. These reporter analyses confirmed that development of the isthmic organizer is regulated by PouV through direct regulation of pax2/pax2a in vertebrate embryos.
Collapse
Affiliation(s)
- Masato Maekawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Shinji Saito
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Institute for Vaccine Research and Development, Hokkaido University, N21, W11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Daiki Isobe
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kazumasa Takemoto
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Road, U3156, Storrs, CT 06269, USA
| | - Yuhei Miura
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Yurie Dobashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| |
Collapse
|
3
|
Li Y, Zhu M, Chen WX, Luo J, Li X, Cao Y, Zheng M, Ma S, Xiao Z, Zhang Y, Jiang L, Wang X, Tan T, Li X, Gong Q, Xiong X, Wang J, Tang M, Li M, Tang YP. A novel mutation in intron 1 of Wnt1 causes developmental loss of dopaminergic neurons in midbrain and ASD-like behaviors in rats. Mol Psychiatry 2023; 28:3795-3805. [PMID: 37658228 PMCID: PMC10730402 DOI: 10.1038/s41380-023-02223-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders with a strong genetic liability. Despite extensive studies, however, the underlying pathogenic mechanism still remains elusive. In the present study, we identified a homozygous mutation in the intron 1 of Wnt1 via large-scale screening of ASD risk/causative genes and verified that this mutation created a new splicing donor site in the intron 1, and consequently, a decrease of WNT1 expression. Interestingly, humanized rat models harboring this mutation exhibited robust ASD-like behaviors including impaired ultrasonic vocalization (USV), decreased social interactions, and restricted and repetitive behaviors. Moreover, in the substantia nigra compacta (SNpc) and the ventral tegmental area (VTA) of mutant rats, dopaminergic (DAergic) neurons were dramatically lost, together with a comparable decrease in striatal DAergic fibers. Furthermore, using single-cell RNA sequencing, we demonstrated that the decreased DAergic neurons in these midbrain areas might attribute to a shift of the boundary of the local pool of progenitor cells from the hypothalamic floor plate to the midbrain floor plate during the early embryonic stage. Moreover, treatments of mutant rats with levodopa could attenuate the impaired USV and social interactions almost completely, but not the restricted and repetitive behaviors. Our results for the first time documented that the developmental loss of DAergic neurons in the midbrain underlies the pathogenesis of ASD, and that the abnormal progenitor cell patterning is a cellular underpinning for this developmental DAergic neuronal loss. Importantly, the effective dopamine therapy suggests a translational significance in the treatment of ASD.
Collapse
Affiliation(s)
- Yongyi Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jing Luo
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yangyang Cao
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Meng Zheng
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhilan Xiao
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yani Zhang
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Linyan Jiang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiumin Wang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ting Tan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xia Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qian Gong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoli Xiong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jun Wang
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mingxi Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Ya-Ping Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
4
|
Zhou J, Yang YJ, Gan RH, Wang Y, Li Z, Zhang XJ, Gui JF, Zhou L. Foxl2a and Foxl2b are involved in midbrain-hindbrain boundary development in zebrafish. Gene Expr Patterns 2022; 46:119286. [PMID: 36341978 DOI: 10.1016/j.gep.2022.119286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/23/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
Foxl2 plays conserved central function in ovarian differentiation and maintenance in several fish species. However, its expression pattern and function in fish embryogenesis are still largely unknown. In this study, we first presented a sequential expression pattern of zebrafish foxl2a and foxl2b during embryo development. They were predominantly expressed in the cranial paraxial mesoderm (CPM) and cranial venous vasculature (CVV) during somitogenesis and subsequently expressed in the pharyngeal arches after 48 h post-fertilization (hpf). Then, we compared the brain structures among zebrafish wildtype (WT) and three homozygous foxl2 mutants (foxl2a-/-, foxl2b-/- and foxl2a-/-;foxl2b-/-) and found the reduction of the fourth ventricle in the three foxl2 mutants, especially in foxl2a-/-;foxl2b-/- mutant. Finally, we detected several key transcription factors involved in the gene regulatory network of midbrain-hindbrain boundary (MHB) patterning, such as wnt1, en1b and pax2a. Their expression levels were obviously downregulated in MHB of foxl2a-/- and foxl2a-/-;foxl2b-/- mutants. Thus, we suggest that Foxl2a and Foxl2b are involved in MHB and the fourth ventricle development in zebrafish. The current study provides insights into the molecular mechanism underlying development of brain ventricular system.
Collapse
Affiliation(s)
- Jian Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Jing Yang
- College of Fisheries, Tianjin Agricultural University, China
| | - Rui-Hai Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Juan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
5
|
Peng L, Zhao Y. Genome-Wide Identification and Expression Profiling of the Wnt Gene Family in Three Rice Planthoppers: Sogatella furcifera, Laodelphax striatellus, and Nilaparvata lugens. JOURNAL OF INSECT SCIENCE (ONLINE) 2022; 22:2. [PMID: 36082678 PMCID: PMC9459440 DOI: 10.1093/jisesa/ieac049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Indexed: 06/15/2023]
Abstract
The Wnt gene family plays essential roles in regulating many developmental processes, including the maintenance of stem cells, cell division, and cell migration. The number of Wnt genes varies among species. Due to the diversity and importance of their functions, the Wnt gene family has gained extensive research interest in various animal species from invertebrates to vertebrates. However, knowledge of the Wnt gene family is limited in rice planthoppers. Three planthopper species, the white-backed planthopper (Sogatella furcifera Horvath), the small brown planthopper (Laodelphax striatellus Fallén) and the brown planthopper (Nilaparvata lugens Stål) (Hemiptera: Delphacidae), are devastating specialist pests of rice and cause serious damage to rice plants. To better study the evolution and function of the Wnt gene family in rice planthoppers, we identified 8 Wnt family genes in three rice planthoppers with both genomic and extensive transcriptomic resources available. We conducted a systematic analysis of the three kinds of rice planthoppers and analyzed the dynamic patterns of gene conservation, as well as Wnt gene loss and duplication. The expression profiles in different developmental stages of S. furcifera and different adult organs and tissues of L. striatellus provide preliminary functional implications for the Wnt genes in rice planthopper. This study presents the first genome-wide study of the Wnt gene family in rice planthoppers, and our findings provide insights into Wnt function and evolution in rice planthoppers.
Collapse
Affiliation(s)
- Lei Peng
- College of Life Science, Guizhou Normal University, Guiyang, China
| | - Yan Zhao
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Hidalgo-Sánchez M, Andreu-Cervera A, Villa-Carballar S, Echevarria D. An Update on the Molecular Mechanism of the Vertebrate Isthmic Organizer Development in the Context of the Neuromeric Model. Front Neuroanat 2022; 16:826976. [PMID: 35401126 PMCID: PMC8987131 DOI: 10.3389/fnana.2022.826976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
A crucial event during the development of the central nervous system (CNS) is the early subdivision of the neural tube along its anterior-to-posterior axis to form neuromeres, morphogenetic units separated by transversal constrictions and programed for particular genetic cascades. The narrower portions observed in the developing neural tube are responsible for relevant cellular and molecular processes, such as clonal restrictions, expression of specific regulatory genes, and differential fate specification, as well as inductive activities. In this developmental context, the gradual formation of the midbrain-hindbrain (MH) constriction has been an excellent model to study the specification of two major subdivisions of the CNS containing the mesencephalic and isthmo-cerebellar primordia. This MH boundary is coincident with the common Otx2-(midbrain)/Gbx2-(hindbrain) expressing border. The early interactions between these two pre-specified areas confer positional identities and induce the generation of specific diffusible morphogenes at this interface, in particular FGF8 and WNT1. These signaling pathways are responsible for the gradual histogenetic specifications and cellular identity acquisitions with in the MH domain. This review is focused on the cellular and molecular mechanisms involved in the specification of the midbrain/hindbrain territory and the formation of the isthmic organizer. Emphasis will be placed on the chick/quail chimeric experiments leading to the acquisition of the first fate mapping and experimental data to, in this way, better understand pioneering morphological studies and innovative gain/loss-of-function analysis.
Collapse
Affiliation(s)
- Matías Hidalgo-Sánchez
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- *Correspondence: Matías Hidalgo-Sánchez Diego Echevarria
| | - Abraham Andreu-Cervera
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Sergio Villa-Carballar
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Diego Echevarria
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
- *Correspondence: Matías Hidalgo-Sánchez Diego Echevarria
| |
Collapse
|
7
|
Said NM, Yassin F, Elkreem EA. Wnt10a missense gene polymorphism association with obesity risk: List of literature and a case-control study with Roc analysis for serum β-catenin level in Egypt. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
8
|
Wahlin KJ, Cheng J, Jurlina SL, Jones MK, Dash NR, Ogata A, Kibria N, Ray S, Eldred KC, Kim C, Heng JS, Phillips J, Johnston RJ, Gamm DM, Berlinicke C, Zack DJ. CRISPR Generated SIX6 and POU4F2 Reporters Allow Identification of Brain and Optic Transcriptional Differences in Human PSC-Derived Organoids. Front Cell Dev Biol 2021; 9:764725. [PMID: 34869356 PMCID: PMC8635054 DOI: 10.3389/fcell.2021.764725] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/11/2021] [Indexed: 01/29/2023] Open
Abstract
Human pluripotent stem cells (PSCs) represent a powerful tool to investigate human eye development and disease. When grown in 3D, they can self-assemble into laminar organized retinas; however, variation in the size, shape and composition of individual organoids exists. Neither the microenvironment nor the timing of critical growth factors driving retinogenesis are fully understood. To explore early retinal development, we developed a SIX6-GFP reporter that enabled the systematic optimization of conditions that promote optic vesicle formation. We demonstrated that early hypoxic growth conditions enhanced SIX6 expression and promoted eye formation. SIX6 expression was further enhanced by sequential inhibition of Wnt and activation of sonic hedgehog signaling. SIX6 + optic vesicles showed RNA expression profiles that were consistent with a retinal identity; however, ventral diencephalic markers were also present. To demonstrate that optic vesicles lead to bona fide "retina-like" structures we generated a SIX6-GFP/POU4F2-tdTomato dual reporter line that labeled the entire developing retina and retinal ganglion cells, respectively. Additional brain regions, including the hypothalamus and midbrain-hindbrain (MBHB) territories were identified by harvesting SIX6 + /POU4F2- and SIX6- organoids, respectively. Using RNAseq to study transcriptional profiles we demonstrated that SIX6-GFP and POU4F2-tdTomato reporters provided a reliable readout for developing human retina, hypothalamus, and midbrain/hindbrain organoids.
Collapse
Affiliation(s)
- Karl J. Wahlin
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States,*Correspondence: Karl J. Wahlin,
| | - Jie Cheng
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shawna L. Jurlina
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Melissa K. Jones
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Nicholas R. Dash
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Anna Ogata
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Nawal Kibria
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Sunayan Ray
- Shiley Eye Institute, University of California, San Diego, San Diego, CA, United States
| | - Kiara C. Eldred
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Catherine Kim
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jacob S. Heng
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, United States
| | - Jenny Phillips
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Robert J. Johnston
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David M. Gamm
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States,Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Cynthia Berlinicke
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Donald J. Zack
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Chang CY, Ting HC, Liu CA, Su HL, Chiou TW, Harn HJ, Lin SZ, Ho TJ. Differentiation of Human Pluripotent Stem Cells Into Specific Neural Lineages. Cell Transplant 2021; 30:9636897211017829. [PMID: 34665040 PMCID: PMC8529300 DOI: 10.1177/09636897211017829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are sources of several somatic cell
types for human developmental studies, in vitro disease modeling, and
cell transplantation therapy. Improving strategies of derivation of
high-purity specific neural and glial lineages from hPSCs is critical
for application to the study and therapy of the nervous system. Here,
we will focus on the principles behind establishment of neuron and
glia differentiation methods according to developmental studies. We
will also highlight the limitations and challenges associated with the
differentiation of several “difficult” neural lineages and delay in
neuronal maturation and functional integration. To overcome these
challenges, we will introduce strategies and novel technologies aimed
at improving the differentiation of various neural lineages to expand
the application potential of hPSCs to the study of the nervous
system.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hsiao-Chien Ting
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Ann Liu
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, Hualien, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Pathology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
10
|
Kesavan G, Machate A, Hans S, Brand M. Cell-fate plasticity, adhesion and cell sorting complementarily establish a sharp midbrain-hindbrain boundary. Development 2020; 147:dev186882. [PMID: 32439756 DOI: 10.1242/dev.186882] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/30/2020] [Indexed: 01/22/2023]
Abstract
The formation and maintenance of sharp boundaries between groups of cells play a vital role during embryonic development as they serve to compartmentalize cells with similar fates. Some of these boundaries also act as organizers, with the ability to induce specific cell fates and morphogenesis in the surrounding cells. The midbrain-hindbrain boundary (MHB) is such an organizer: it acts as a lineage restriction boundary to prevent the intermingling of cells with different developmental fates. However, the mechanisms underlying the lineage restriction process remain unclear. Here, using novel fluorescent knock-in reporters, live imaging, Cre/lox-mediated lineage tracing, atomic force microscopy-based cell adhesion assays and mutant analysis, we analyze the process of lineage restriction at the MHB and provide mechanistic details. Specifically, we show that lineage restriction occurs by the end of gastrulation, and that the subsequent formation of sharp gene expression boundaries in the developing MHB occur through complementary mechanisms, i.e. cell-fate plasticity and cell sorting. Furthermore, we show that cell sorting at the MHB involves differential adhesion among midbrain and hindbrain cells that is mediated by N-cadherin and Eph-ephrin signaling.
Collapse
Affiliation(s)
- Gokul Kesavan
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Anja Machate
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| |
Collapse
|
11
|
Induced Pluripotent Stem Cell (iPSC)-Based Neurodegenerative Disease Models for Phenotype Recapitulation and Drug Screening. Molecules 2020; 25:molecules25082000. [PMID: 32344649 PMCID: PMC7221979 DOI: 10.3390/molecules25082000] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases represent a significant unmet medical need in our aging society. There are no effective treatments for most of these diseases, and we know comparatively little regarding pathogenic mechanisms. Among the challenges faced by those involved in developing therapeutic drugs for neurodegenerative diseases, the syndromes are often complex, and small animal models do not fully recapitulate the unique features of the human nervous system. Human induced pluripotent stem cells (iPSCs) are a novel technology that ideally would permit us to generate neuronal cells from individual patients, thereby eliminating the problem of species-specificity inherent when using animal models. Specific phenotypes of iPSC-derived cells may permit researchers to identify sub-types and to distinguish among unique clusters and groups. Recently, iPSCs were used for drug screening and testing for neurologic disorders including Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), spinocerebellar atrophy (SCA), and Zika virus infection. However, there remain many challenges still ahead, including how one might effectively recapitulate sporadic disease phenotypes and the selection of ideal phenotypes and for large-scale drug screening. Fortunately, quite a few novel strategies have been developed that might be combined with an iPSC-based model to solve these challenges, including organoid technology, single-cell RNA sequencing, genome editing, and deep learning artificial intelligence. Here, we will review current applications and potential future directions for iPSC-based neurodegenerative disease models for critical drug screening.
Collapse
|
12
|
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
|
13
|
Frank D, Sela-Donenfeld D. Hindbrain induction and patterning during early vertebrate development. Cell Mol Life Sci 2019; 76:941-960. [PMID: 30519881 PMCID: PMC11105337 DOI: 10.1007/s00018-018-2974-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/28/2022]
Abstract
The hindbrain is a key relay hub of the central nervous system (CNS), linking the bilaterally symmetric half-sides of lower and upper CNS centers via an extensive network of neural pathways. Dedicated neural assemblies within the hindbrain control many physiological processes, including respiration, blood pressure, motor coordination and different sensations. During early development, the hindbrain forms metameric segmented units known as rhombomeres along the antero-posterior (AP) axis of the nervous system. These compartmentalized units are highly conserved during vertebrate evolution and act as the template for adult brainstem structure and function. TALE and HOX homeodomain family transcription factors play a key role in the initial induction of the hindbrain and its specification into rhombomeric cell fate identities along the AP axis. Signaling pathways, such as canonical-Wnt, FGF and retinoic acid, play multiple roles to initially induce the hindbrain and regulate Hox gene-family expression to control rhombomeric identity. Additional transcription factors including Krox20, Kreisler and others act both upstream and downstream to Hox genes, modulating their expression and protein activity. In this review, we will examine the earliest embryonic signaling pathways that induce the hindbrain and subsequent rhombomeric segmentation via Hox and other gene expression. We will examine how these signaling pathways and transcription factors interact to activate downstream targets that organize the segmented AP pattern of the embryonic vertebrate hindbrain.
Collapse
Affiliation(s)
- Dale Frank
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, 31096, Haifa, Israel.
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel.
| |
Collapse
|
14
|
Lekven AC, Lilie CJ, Gibbs HC, Green DG, Singh A, Yeh AT. Analysis of the wnt1 regulatory chromosomal landscape. Dev Genes Evol 2019; 229:43-52. [PMID: 30825002 DOI: 10.1007/s00427-019-00629-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022]
Abstract
One of the earliest patterning events in the vertebrate neural plate is the specification of mes/r1, the territory comprising the prospective mesencephalon and the first hindbrain rhombomere. Within mes/r1, an interface of gene expression defines the midbrain-hindbrain boundary (MHB), a lineage restriction that separates the mesencephalon and rhombencephalon. wnt1 is critical to mes/r1 development and functions within the MHB as a component of the MHB gene regulatory network (GRN). Despite its importance to these critical and early steps of vertebrate neurogenesis, little is known about the factors responsible for wnt1 transcriptional regulation. In the zebrafish, wnt1 and its neighboring paralog, wnt10b, are expressed in largely overlapping patterns, suggesting co-regulation. To understand wnt1 and wnt10b transcriptional control, we used a comparative genomics approach to identify relevant enhancers. We show that the wnt1-wnt10b locus contains multiple cis-regulatory elements that likely interact to generate the wnt1 and wnt10b expression patterns. Two of 11 conserved enhancers tested show activity restricted to the midbrain and MHB, an activity that is conserved in the distantly related spotted gar orthologous elements. Three non-conserved elements also play a likely role in wnt1 regulation. The identified enhancers display dynamic modes of chromatin accessibility, suggesting controlled deployment during embryogenesis. Our results suggest that the control of wnt1 and wnt10b expression is under complex regulation involving the interaction of multiple enhancers.
Collapse
Affiliation(s)
- Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA. .,Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA.
| | - Craig J Lilie
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - David G Green
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Avantika Singh
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| |
Collapse
|
15
|
Somorjai IML, Martí-Solans J, Diaz-Gracia M, Nishida H, Imai KS, Escrivà H, Cañestro C, Albalat R. Wnt evolution and function shuffling in liberal and conservative chordate genomes. Genome Biol 2018; 19:98. [PMID: 30045756 PMCID: PMC6060547 DOI: 10.1186/s13059-018-1468-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND What impact gene loss has on the evolution of developmental processes, and how function shuffling has affected retained genes driving essential biological processes, remain open questions in the fields of genome evolution and EvoDevo. To investigate these problems, we have analyzed the evolution of the Wnt ligand repertoire in the chordate phylum as a case study. RESULTS We conduct an exhaustive survey of Wnt genes in genomic databases, identifying 156 Wnt genes in 13 non-vertebrate chordates. This represents the most complete Wnt gene catalog of the chordate subphyla and has allowed us to resolve previous ambiguities about the orthology of many Wnt genes, including the identification of WntA for the first time in chordates. Moreover, we create the first complete expression atlas for the Wnt family during amphioxus development, providing a useful resource to investigate the evolution of Wnt expression throughout the radiation of chordates. CONCLUSIONS Our data underscore extraordinary genomic stasis in cephalochordates, which contrasts with the liberal and dynamic evolutionary patterns of gene loss and duplication in urochordate genomes. Our analysis has allowed us to infer ancestral Wnt functions shared among all chordates, several cases of function shuffling among Wnt paralogs, as well as unique expression domains for Wnt genes that likely reflect functional innovations in each chordate lineage. Finally, we propose a potential relationship between the evolution of WntA and the evolution of the mouth in chordates.
Collapse
Affiliation(s)
- Ildikó M L Somorjai
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9ST, Scotland, UK.
- Scottish Oceans Institute, School of Biology, University of St Andrews, East Sands, St Andrews, KY16 8LB, Scotland, UK.
| | - Josep Martí-Solans
- Departament de Genètica, , Microbiologia i Estadística, and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain
| | - Miriam Diaz-Gracia
- Departament de Genètica, , Microbiologia i Estadística, and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain
| | - Hiroki Nishida
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, 560-0043, Japan
| | - Kaoru S Imai
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, 560-0043, Japan
| | - Hector Escrivà
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie Intégrative des Organismes Marins (BIOM), Observatoire Océanologique, F-66650, Banyuls/Mer, France
| | - Cristian Cañestro
- Departament de Genètica, , Microbiologia i Estadística, and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain.
| | - Ricard Albalat
- Departament de Genètica, , Microbiologia i Estadística, and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
16
|
Lindsey BW, Hall ZJ, Heuzé A, Joly JS, Tropepe V, Kaslin J. The role of neuro-epithelial-like and radial-glial stem and progenitor cells in development, plasticity, and repair. Prog Neurobiol 2018; 170:99-114. [PMID: 29902500 DOI: 10.1016/j.pneurobio.2018.06.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/20/2018] [Accepted: 06/07/2018] [Indexed: 12/14/2022]
Abstract
Neural stem and progenitor cells (NSPCs) are the primary source of new neurons in the brain and serve critical roles in tissue homeostasis and plasticity throughout life. Within the vertebrate brain, NSPCs are located within distinct neurogenic niches differing in their location, cellular composition, and proliferative behaviour. Heterogeneity in the NSPC population is hypothesized to reflect varying capacities for neurogenesis, plasticity and repair between different neurogenic zones. Since the discovery of adult neurogenesis, studies have predominantly focused on the behaviour and biological significance of adult NSPCs (aNSPCs) in rodents. However, compared to rodents, who show lifelong neurogenesis in only two restricted neurogenic niches, zebrafish exhibit constitutive neurogenesis across multiple stem cell niches that provide new neurons to every major brain division. Accordingly, zebrafish are a powerful model to probe the unique cellular and molecular profiles of NSPCs and investigate how these profiles govern tissue homeostasis and regenerative plasticity within distinct stem cell populations over time. Amongst the NSPC populations residing in the zebrafish central nervous system (CNS), proliferating radial-glia, quiescent radial-glia and neuro-epithelial-like cells comprise the majority. Here, we provide insight into the extent to which these distinct NSPC populations function and mature during development, respond to experience, and contribute to successful CNS regeneration in teleost fish. Together, our review brings to light the dynamic biological roles of these individual NSPC populations and showcases their diverse regenerative modes to achieve vertebrate brain repair later in life.
Collapse
Affiliation(s)
- Benjamin W Lindsey
- Department of Biology, Brain and Mind Research Institute, University of Ottawa, Ontario, Canada; Australian Regenerative Medicine Institute, Monash University Clayton Campus, Clayton, VIC, Australia.
| | - Zachary J Hall
- Department of Cell and Systems Biology, University of Toronto, Ontario, M5S 3G5, Canada.
| | - Aurélie Heuzé
- CASBAH INRA group, UMR9197 Neuro-PSI, CNRS, 91 198, Gif-sur-Yvette, France.
| | - Jean-Stéphane Joly
- CASBAH INRA group, UMR9197 Neuro-PSI, CNRS, 91 198, Gif-sur-Yvette, France.
| | - Vincent Tropepe
- Department of Cell and Systems Biology, University of Toronto, Ontario, M5S 3G5, Canada.
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University Clayton Campus, Clayton, VIC, Australia.
| |
Collapse
|
17
|
Silva N, Louro B, Trindade M, Power DM, Campinho MA. Transcriptomics reveal an integrative role for maternal thyroid hormones during zebrafish embryogenesis. Sci Rep 2017; 7:16657. [PMID: 29192226 PMCID: PMC5709499 DOI: 10.1038/s41598-017-16951-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023] Open
Abstract
Thyroid hormones (THs) are essential for embryonic brain development but the genetic mechanisms involved in the action of maternal THs (MTHs) are still largely unknown. As the basis for understanding the underlying genetic mechanisms of MTHs regulation we used an established zebrafish monocarboxylic acid transporter 8 (MCT8) knock-down model and characterised the transcriptome in 25hpf zebrafish embryos. Subsequent mapping of differentially expressed genes using Reactome pathway analysis together with in situ expression analysis and immunohistochemistry revealed the genetic networks and cells under MTHs regulation during zebrafish embryogenesis. We found 4,343 differentially expressed genes and the Reactome pathway analysis revealed that TH is involved in 1681 of these pathways. MTHs regulated the expression of core developmental pathways, such as NOTCH and WNT in a cell specific context. The cellular distribution of neural MTH-target genes demonstrated their cell specific action on neural stem cells and differentiated neuron classes. Taken together our data show that MTHs have a role in zebrafish neurogenesis and suggest they may be involved in cross talk between key pathways in neural development. Given that the observed MCT8 zebrafish knockdown phenotype resembles the symptoms in human patients with Allan-Herndon-Dudley syndrome our data open a window into understanding the genetics of this human congenital condition.
Collapse
Affiliation(s)
- Nadia Silva
- Comparative Endocrinology and Integrative Biology Group, Centre for Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Bruno Louro
- Comparative Endocrinology and Integrative Biology Group, Centre for Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Marlene Trindade
- Comparative Endocrinology and Integrative Biology Group, Centre for Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Deborah M Power
- Comparative Endocrinology and Integrative Biology Group, Centre for Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Marco A Campinho
- Comparative Endocrinology and Integrative Biology Group, Centre for Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal.
| |
Collapse
|
18
|
Gibbs HC, Chang-Gonzalez A, Hwang W, Yeh AT, Lekven AC. Midbrain-Hindbrain Boundary Morphogenesis: At the Intersection of Wnt and Fgf Signaling. Front Neuroanat 2017; 11:64. [PMID: 28824384 PMCID: PMC5541008 DOI: 10.3389/fnana.2017.00064] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
A constriction in the neural tube at the junction of the midbrain and hindbrain is a conserved feature of vertebrate embryos. The constriction is a defining feature of the midbrain-hindbrain boundary (MHB), a signaling center that patterns the adjacent midbrain and rostral hindbrain and forms at the junction of two gene expression domains in the early neural plate: an anterior otx2/wnt1 positive domain and a posterior gbx/fgf8 positive domain. otx2 and gbx genes encode mutually repressive transcription factors that create a lineage restriction boundary at their expression interface. Wnt and Fgf genes form a mutually dependent feedback system that maintains their expression domains on the otx2 or gbx side of the boundary, respectively. Constriction morphogenesis occurs after these conserved gene expression domains are established and while their mutual interactions maintain their expression pattern; consequently, mutant studies in zebrafish have led to the suggestion that constriction morphogenesis should be considered a unique phase of MHB development. We analyzed MHB morphogenesis in fgf8 loss of function zebrafish embryos using a reporter driven by the conserved wnt1 enhancer to visualize anterior boundary cells. We found that fgf8 loss of function results in a re-activation of wnt1 reporter expression posterior to the boundary simultaneous with an inactivation of the wnt1 reporter in the anterior boundary cells, and that these events correlate with relaxation of the boundary constriction. In consideration of other results that correlate the boundary constriction with Wnt and Fgf expression, we propose that the maintenance of an active Wnt-Fgf feedback loop is a key factor in driving the morphogenesis of the MHB constriction.
Collapse
Affiliation(s)
- Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Ana Chang-Gonzalez
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Wonmuk Hwang
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States.,Department of Materials Science and Engineering, Texas A&M UniversityCollege Station, TX, United States.,School of Computational Sciences, Korea Institute for Advanced StudySeoul, South Korea
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M UniversityCollege Station, TX, United States
| | - Arne C Lekven
- Department of Biology, Texas A&M UniversityCollege Station, TX, United States
| |
Collapse
|
19
|
Shitasako S, Ito Y, Ito R, Ueda Y, Shimizu Y, Ohshima T. Wnt and Shh signals regulate neural stem cell proliferation and differentiation in the optic tectum of adult zebrafish. Dev Neurobiol 2017; 77:1206-1220. [DOI: 10.1002/dneu.22509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/29/2017] [Accepted: 06/02/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Shiori Shitasako
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| | - Yoko Ito
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| | - Ryoichi Ito
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| | - Yuto Ueda
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| | - Yuki Shimizu
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science; Waseda University, 2-2 Wakamatsu-cho; Shinjuku-ku Tokyo 162-8480 Japan
| |
Collapse
|
20
|
Cheung TT, Weston MK, Wilson MJ. Selection and evaluation of reference genes for analysis of mouse (Mus musculus) sex-dimorphic brain development. PeerJ 2017; 5:e2909. [PMID: 28133578 PMCID: PMC5251938 DOI: 10.7717/peerj.2909] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/08/2016] [Indexed: 11/24/2022] Open
Abstract
The development of the brain is sex-dimorphic, and as a result so are many neurological disorders. One approach for studying sex-dimorphic brain development is to measure gene expression in biological samples using RT-qPCR. However, the accuracy and consistency of this technique relies on the reference gene(s) selected. We analyzed the expression of ten reference genes in male and female samples over three stages of brain development, using popular algorithms NormFinder, GeNorm and Bestkeeper. The top ranked reference genes at each time point were further used to quantify gene expression of three sex-dimorphic genes (Wnt10b, Xist and CYP7B1). When comparing gene expression between the sexes expression at specific time points the best reference gene combinations are: Sdha/Pgk1 at E11.5, RpL38/Sdha E12.5, and Actb/RpL37 at E15.5. When studying expression across time, the ideal reference gene(s) differs with sex. For XY samples a combination of Actb/Sdha. In contrast, when studying gene expression across developmental stage with XX samples, Sdha/Gapdh were the top reference genes. Our results identify the best combination of two reference genes when studying male and female brain development, and emphasize the importance of selecting the correct reference genes for comparisons between developmental stages.
Collapse
Affiliation(s)
- Tanya T Cheung
- Department of Anatomy, University of Otago , Dunedin , New Zealand
| | | | - Megan J Wilson
- Department of Anatomy, University of Otago , Dunedin , New Zealand
| |
Collapse
|
21
|
Liu H, Liu Q, Zhou X, Huang Y, Zhang Z. Genome Editing of Wnt-1, a Gene Associated with Segmentation, via CRISPR/Cas9 in the Pine Caterpillar Moth, Dendrolimus punctatus. Front Physiol 2017; 7:666. [PMID: 28111552 PMCID: PMC5216022 DOI: 10.3389/fphys.2016.00666] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 12/19/2016] [Indexed: 01/11/2023] Open
Abstract
The pine caterpillar moth, Dendrolimus punctatus, is a devastating forest pest. Genetic manipulation of this insect pest is limited due to the lack of genomic and functional genomic toolsets. Recently, CRISPR/Cas9 technology has been demonstrated to be a promising approach to modify the genome. To investigate gene functions during the embryogenesis, we introduced CRISPR/Cas9 system in D. punctatus to precisely and effectively manipulate gene expressions inmutant embryos. Compared to controls, knocking out of DpWnt-1, a gene well known for its role in the early body planning, led to high embryonic mortality. Among these mutants, 32.9% of the embryos and larvae showed an abnormal development. DpWnt-1 mutants predominantly exhibited abnormal posterior segments. In addition, multiple phenotypes were observed, including the loss of limbs and the head deformation, suggesting that DpWnt-1 signaling pathway is necessary for anterior segmentation and appendage development. Overall, our results demonstrate that CRISPR/Cas9 system is feasible and efficient in inducing mutations at a specific locus in D. punctatus. This study not only lays the foundation for characterizing gene functions in a non-model species, but also facilitates the future development of pest control alternatives for a major defoliator.
Collapse
Affiliation(s)
- Huihui Liu
- Key Laboratory of Forest Protection, Research Institute of Forest Ecology, Environment and Protection, Chinese Academy of Forestry, State Forestry Administration Beijing, China
| | - Qun Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences Shanghai, China
| | - Xuguo Zhou
- Department of Entomology, University of Kentucky Lexington, KY, USA
| | - Yongping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences Shanghai, China
| | - Zhen Zhang
- Key Laboratory of Forest Protection, Research Institute of Forest Ecology, Environment and Protection, Chinese Academy of Forestry, State Forestry Administration Beijing, China
| |
Collapse
|
22
|
Peretz Y, Eren N, Kohl A, Hen G, Yaniv K, Weisinger K, Cinnamon Y, Sela-Donenfeld D. A new role of hindbrain boundaries as pools of neural stem/progenitor cells regulated by Sox2. BMC Biol 2016; 14:57. [PMID: 27392568 PMCID: PMC4938926 DOI: 10.1186/s12915-016-0277-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/21/2016] [Indexed: 01/28/2023] Open
Abstract
Background Compartment boundaries are an essential developmental mechanism throughout evolution, designated to act as organizing centers and to regulate and localize differently fated cells. The hindbrain serves as a fascinating example for this phenomenon as its early development is devoted to the formation of repetitive rhombomeres and their well-defined boundaries in all vertebrates. Yet, the actual role of hindbrain boundaries remains unresolved, especially in amniotes. Results Here, we report that hindbrain boundaries in the chick embryo consist of a subset of cells expressing the key neural stem cell (NSC) gene Sox2. These cells co-express other neural progenitor markers such as Transitin (the avian Nestin), GFAP, Pax6 and chondroitin sulfate proteoglycan. The majority of the Sox2+ cells that reside within the boundary core are slow-dividing, whereas nearer to and within rhombomeres Sox2+ cells are largely proliferating. In vivo analyses and cell tracing experiments revealed the contribution of boundary Sox2+ cells to neurons in a ventricular-to-mantle manner within the boundaries, as well as their lateral contribution to proliferating Sox2+ cells in rhombomeres. The generation of boundary-derived neurospheres from hindbrain cultures confirmed the typical NSC behavior of boundary cells as a multipotent and self-renewing Sox2+ cell population. Inhibition of Sox2 in boundaries led to enhanced and aberrant neural differentiation together with inhibition in cell-proliferation, whereas Sox2 mis-expression attenuated neurogenesis, confirming its significant function in hindbrain neuronal organization. Conclusions Data obtained in this study deciphers a novel role of hindbrain boundaries as repetitive pools of neural stem/progenitor cells, which provide proliferating progenitors and differentiating neurons in a Sox2-dependent regulation. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0277-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuval Peretz
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Noa Eren
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Gideon Hen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karen Weisinger
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Yuval Cinnamon
- Institute of Animal Sciences, Department of Poultry and Aquaculture Sciences, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel.
| |
Collapse
|
23
|
Zizioli D, Tiso N, Guglielmi A, Saraceno C, Busolin G, Giuliani R, Khatri D, Monti E, Borsani G, Argenton F, Finazzi D. Knock-down of pantothenate kinase 2 severely affects the development of the nervous and vascular system in zebrafish, providing new insights into PKAN disease. Neurobiol Dis 2015; 85:35-48. [PMID: 26476142 PMCID: PMC4684146 DOI: 10.1016/j.nbd.2015.10.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/31/2015] [Accepted: 10/11/2015] [Indexed: 01/31/2023] Open
Abstract
Pantothenate Kinase Associated Neurodegeneration (PKAN) is an autosomal recessive disorder with mutations in the pantothenate kinase 2 gene (PANK2), encoding an essential enzyme for Coenzyme A (CoA) biosynthesis. The molecular connection between defects in this enzyme and the neurodegenerative phenotype observed in PKAN patients is still poorly understood. We exploited the zebrafish model to study the role played by the pank2 gene during embryonic development and get new insight into PKAN pathogenesis. The zebrafish orthologue of hPANK2 lies on chromosome 13, is a maternal gene expressed in all development stages and, in adult animals, is highly abundant in CNS, dorsal aorta and caudal vein. The injection of a splice-inhibiting morpholino induced a clear phenotype with perturbed brain morphology and hydrocephalus; edema was present in the heart region and caudal plexus, where hemorrhages with reduction of blood circulation velocity were detected. We characterized the CNS phenotype by studying the expression pattern of wnt1 and neurog1 neural markers and by use of the Tg(neurod:EGFP/sox10:dsRed) transgenic line. The results evidenced that downregulation of pank2 severely impairs neuronal development, particularly in the anterior part of CNS (telencephalon). Whole-mount in situ hybridization analysis of the endothelial markers cadherin-5 and fli1a, and use of Tg(fli1a:EGFP/gata1a:dsRed) transgenic line, confirmed the essential role of pank2 in the formation of the vascular system. The specificity of the morpholino-induced phenotype was proved by the restoration of a normal development in a high percentage of embryos co-injected with pank2 mRNA. Also, addition of pantethine or CoA, but not of vitamin B5, to pank2 morpholino-injected embryos rescued the phenotype with high efficiency. The zebrafish model indicates the relevance of pank2 activity and CoA homeostasis for normal neuronal development and functioning and provides evidence of an unsuspected role for this enzyme and its product in vascular development. Zebrafish pank2 gene is highly expressed in the CNS and the main vascular structures. Pank2 down-regulation severely affects the development of the forebrain. Pank2 down-regulation affects the dorsal aorta, caudal vein and inter-somitic vessels. Pantethine and Coenzyme A restore the normal development in the absence of pank2 expression.
Collapse
Affiliation(s)
- Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy.
| | - Natascia Tiso
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Adele Guglielmi
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Claudia Saraceno
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Giorgia Busolin
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Roberta Giuliani
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Deepak Khatri
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Giuseppe Borsani
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Francesco Argenton
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Dario Finazzi
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy; Clinical Chemistry Laboratory, Spedali Civili Hospital, 25123 Brescia, Italy.
| |
Collapse
|
24
|
slc7a6os gene plays a critical role in defined areas of the developing CNS in zebrafish. PLoS One 2015; 10:e0119696. [PMID: 25803583 PMCID: PMC4372478 DOI: 10.1371/journal.pone.0119696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/15/2015] [Indexed: 11/19/2022] Open
Abstract
The aim of this study is to shed light on the functional role of slc7a6os, a gene highly conserved in vertebrates. The Danio rerio slc7a6os gene encodes a protein of 326 amino acids with 46% identity to human SLC7A6OS and 14% to Saccharomyces cerevisiae polypeptide Iwr1. Yeast Iwr1 specifically binds RNA pol II, interacts with the basal transcription machinery and regulates the transcription of specific genes. In this study we investigated for the first time the biological role of SLC7A6OS in vertebrates. Zebrafish slc7a6os is a maternal gene that is expressed throughout development, with a prevalent localization in the developing central nervous system (CNS). The gene is also expressed, although at different levels, in various tissues of the adult fish. To determine the functional role of slc7a6os during zebrafish development, we knocked-down the gene by injecting a splice-blocking morpholino. At 24 hpf morphants show morphological defects in the CNS, particularly the interface between hindbrain and midbrain is not well-defined. At 28 hpf the morpholino injected embryos present an altered somite morphology and appear partially or completely immotile. At this stage the midbrain, hindbrain and cerebellum are compromised and not well defined compared with control embryos. The observed alterations persist at later developmental stages. Consistently, the expression pattern of two markers specifically expressed in the developing CNS, pax2a and neurod, is significantly altered in morphants. The co-injection of embryos with synthetic slc7a6os mRNA, rescues the morphant phenotype and restores the wild type expression pattern of pax2a and neurod. Our data suggest that slc7a6os might play a critical role in defined areas of the developing CNS in vertebrates, probably by regulating the expression of key genes.
Collapse
|
25
|
Pose-Méndez S, Candal E, Mazan S, Rodríguez-Moldes I. Genoarchitecture of the rostral hindbrain of a shark: basis for understanding the emergence of the cerebellum at the agnathan–gnathostome transition. Brain Struct Funct 2015; 221:1321-35. [DOI: 10.1007/s00429-014-0973-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 12/17/2014] [Indexed: 12/14/2022]
|
26
|
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
|
27
|
Zizioli D, Guarienti M, Tobia C, Gariano G, Borsani G, Bresciani R, Ronca R, Giacopuzzi E, Preti A, Gaudenzi G, Belleri M, Di Salle E, Fabrias G, Casas J, Ribatti D, Monti E, Presta M. Molecular cloning and knockdown of galactocerebrosidase in zebrafish: New insights into the pathogenesis of Krabbe's disease. Biochim Biophys Acta Mol Basis Dis 2014; 1842:665-75. [DOI: 10.1016/j.bbadis.2014.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/17/2013] [Accepted: 01/15/2014] [Indexed: 11/30/2022]
|
28
|
Thrasivoulou C, Millar M, Ahmed A. Activation of intracellular calcium by multiple Wnt ligands and translocation of β-catenin into the nucleus: a convergent model of Wnt/Ca2+ and Wnt/β-catenin pathways. J Biol Chem 2013; 288:35651-9. [PMID: 24158438 PMCID: PMC3861617 DOI: 10.1074/jbc.m112.437913] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Ca2+ and β-catenin, a 92-kDa negatively charged transcription factor, transduce Wnt signaling via the non-canonical, Wnt/Ca2+ and canonical, Wnt/β-catenin pathways independently. The nuclear envelope is a barrier to large protein entry, and this process is regulated by intracellular calcium [Ca2+]i and trans-nuclear potential. How β-catenin traverses the nuclear envelope is not well known. We hypothesized that Wnt/Ca2+ and Wnt/β-catenin pathways act in a coordinated manner and that [Ca2+]i release facilitates β-catenin entry into the nucleus in mammalian cells. In a live assay using calcium dyes in PC3 prostate cancer cells, six Wnt peptides (3A, 4, 5A, 7A, 9B, and 10B) mobilized [Ca2+]i but Wnt11 did not. Based upon dwell time (range = 15–30 s) of the calcium waveform, these Wnts could be classified into three classes: short, 3A and 5A; long, 7A and 10B; and very long, 4 and 9B. Wnt-activated [Ca2+]i release was followed by an increase in intranuclear calcium and the depolarization of both the cell and nuclear membranes, determined by using FM4-64. In cells treated with Wnts 5A, 9B, and 10B, paradigm substrates for each Wnt class, increased [Ca2+]i was followed by β-catenin translocation into the nucleus in PC3, MCF7, and 253J, prostate, breast, and bladder cancer cell lines; both the increase in Wnt 5A, 9B, and 10B induced [Ca2+]i release and β-catenin translocation are suppressed by thapsigargin in PC3 cell line. We propose a convergent model of Wnt signaling network where Ca2+ and β-catenin pathways may act in a coordinated, interdependent, rather than independent, manner.
Collapse
Affiliation(s)
- Christopher Thrasivoulou
- From the Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics, Rockefeller Building, University Street, University College London, London WC1E 6JJ, United Kingdom
| | | | | |
Collapse
|
29
|
Cornelia de Lange Syndrome: NIPBL haploinsufficiency downregulates canonical Wnt pathway in zebrafish embryos and patients fibroblasts. Cell Death Dis 2013; 4:e866. [PMID: 24136230 PMCID: PMC3824680 DOI: 10.1038/cddis.2013.371] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 01/21/2023]
Abstract
Cornelia de Lange Syndrome is a severe genetic disorder characterized by malformations affecting multiple systems, with a common feature of severe mental retardation. Genetic variants within four genes (NIPBL (Nipped-B-like), SMC1A, SMC3, and HDAC8) are believed to be responsible for the majority of cases; all these genes encode proteins that are part of the 'cohesin complex'. Cohesins exhibit two temporally separated major roles in cells: one controlling the cell cycle and the other involved in regulating the gene expression. The present study focuses on the role of the zebrafish nipblb paralog during neural development, examining its expression in the central nervous system, and analyzing the consequences of nipblb loss of function. Neural development was impaired by the knockdown of nipblb in zebrafish. nipblb-loss-of-function embryos presented with increased apoptosis in the developing neural tissues, downregulation of canonical Wnt pathway genes, and subsequent decreased Cyclin D1 (Ccnd1) levels. Importantly, the same pattern of canonical WNT pathway and CCND1 downregulation was observed in NIPBL-mutated patient-specific fibroblasts. Finally, chemical activation of the pathway in nipblb-loss-of-function embryos rescued the adverse phenotype and restored the physiological levels of cell death.
Collapse
|
30
|
Schmidt R, Strähle U, Scholpp S. Neurogenesis in zebrafish - from embryo to adult. Neural Dev 2013; 8:3. [PMID: 23433260 PMCID: PMC3598338 DOI: 10.1186/1749-8104-8-3] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/17/2013] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis in the developing central nervous system consists of the induction and proliferation of neural progenitor cells and their subsequent differentiation into mature neurons. External as well as internal cues orchestrate neurogenesis in a precise temporal and spatial way. In the last 20 years, the zebrafish has proven to be an excellent model organism to study neurogenesis in the embryo. Recently, this vertebrate has also become a model for the investigation of adult neurogenesis and neural regeneration. Here, we summarize the contributions of zebrafish in neural development and adult neurogenesis.
Collapse
Affiliation(s)
- Rebecca Schmidt
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76021, Karlsruhe, Germany
| | | | | |
Collapse
|
31
|
Shimizu N, Kawakami K, Ishitani T. Visualization and exploration of Tcf/Lef function using a highly responsive Wnt/β-catenin signaling-reporter transgenic zebrafish. Dev Biol 2012; 370:71-85. [PMID: 22842099 DOI: 10.1016/j.ydbio.2012.07.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 07/03/2012] [Accepted: 07/10/2012] [Indexed: 01/09/2023]
Abstract
Evolutionarily conserved Tcf/Lef transcription factors (Lef1, Tcf7, Tcf7l1, and Tcf7l2) mediate gene expression regulated by Wnt/β-catenin signaling, which has multiple roles in early embryogenesis, organogenesis, adult tissue homeostasis, and tissue regeneration. However, the spatiotemporal dynamics of Tcf/Lef activity during these events remain poorly understood. We generated stable transgenic zebrafish lines carrying a new Wnt/β-catenin signaling reporter, Tcf/Lef-miniP:dGFP. The reporter revealed the transcriptional activities of four Tcf/Lef members controlled by Wnt/β-catenin signaling, which were expressed in known Wnt/β-catenin signaling-active sites during embryogenesis, organ development and growth, and tissue regeneration. We used the transgenic lines to demonstrate the contribution of Tcf/Lef-mediated Wnt/β-catenin signaling to the development of the anterior lateral line, dorsal and secondary posterior lateral lines, and gill filaments. Thus, these reporter lines are highly useful tools for studying Tcf/Lef-mediated Wnt/β-catenin signaling-dependent processes.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Division of Cell Regulation Systems, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | |
Collapse
|
32
|
Mattes B, Weber S, Peres J, Chen Q, Davidson G, Houart C, Scholpp S. Wnt3 and Wnt3a are required for induction of the mid-diencephalic organizer in the caudal forebrain. Neural Dev 2012; 7:12. [PMID: 22475147 PMCID: PMC3349543 DOI: 10.1186/1749-8104-7-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/04/2012] [Indexed: 01/05/2023] Open
Abstract
Background A fundamental requirement for development of diverse brain regions is the function of local organizers at morphological boundaries. These organizers are restricted groups of cells that secrete signaling molecules, which in turn regulate the fate of the adjacent neural tissue. The thalamus is located in the caudal diencephalon and is the central relay station between the sense organs and higher brain areas. The mid-diencephalic organizer (MDO) orchestrates the development of the thalamus by releasing secreted signaling molecules such as Shh. Results Here we show that canonical Wnt signaling in the caudal forebrain is required for the formation of the Shh-secreting MD organizer in zebrafish. Wnt signaling induces the MDO in a narrow time window of 4 hours - between 10 and 14 hours post fertilization. Loss of Wnt3 and Wnt3a prevents induction of the MDO, a phenotype also observed upon blockage of canonical Wnt signaling per se. Pharmaceutical activation of the canonical Wnt pathways in Wnt3/Wnt3a compound morphant embryos is able to restore the lack of the MDO. After blockage of Wnt signaling or knock-down of Wnt3/Wnt3a we find an increase of apoptotic cells specifically within the organizer primordium. Consistently, blockage of apoptosis restores the thalamus organizer MDO in Wnt deficient embryos. Conclusion We have identified canonical Wnt signaling as a novel pathway, that is required for proper formation of the MDO and consequently for the development of the major relay station of the brain - the thalamus. We propose that Wnt ligands are necessary to maintain the primordial tissue of the organizer during somitogenesis by suppressing Tp53-mediated apoptosis.
Collapse
Affiliation(s)
- Benjamin Mattes
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Karlsruhe, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Ota S, Ishitani S, Shimizu N, Matsumoto K, Itoh M, Ishitani T. NLK positively regulates Wnt/β-catenin signalling by phosphorylating LEF1 in neural progenitor cells. EMBO J 2012; 31:1904-15. [PMID: 22373574 DOI: 10.1038/emboj.2012.46] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 01/30/2012] [Indexed: 12/26/2022] Open
Abstract
Nemo-like kinase (NLK/Nlk) is an evolutionarily conserved protein kinase involved in Wnt/β-catenin signalling. However, the roles of NLK in Wnt/β-catenin signalling in vertebrates remain unclear. Here, we show that inhibition of Nlk2 function in zebrafish results in decreased Lymphoid enhancer factor-1 (Lef1)-mediated gene expression and cell proliferation in the presumptive midbrain, resulting in a reduction of midbrain tectum size. These defects are related to phosphorylation of Lef1 by Nlk2. Thus, Nlk2 is essential for the phosphorylation and activation of Lef1 transcriptional activity in neural progenitor cells (NPCs). In NPC-like mammalian cells, NLK is also required for the phosphorylation and activation of LEF1 transcriptional activity. Phosphorylation of LEF1 induces its dissociation from histone deacetylase, thereby allowing transcription activation. Furthermore, we demonstrate that NLK functions downstream of Dishevelled (Dvl) in the Wnt/β-catenin signalling pathway. Our findings reveal a novel role of NLK in the activation of the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Satoshi Ota
- Division of Cell Regulation Systems, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Chen Q, Takada R, Takada S. Loss of Porcupine impairs convergent extension during gastrulation in zebrafish. J Cell Sci 2012; 125:2224-34. [PMID: 22357957 DOI: 10.1242/jcs.098368] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porcupine (Porcn), an O-acyltransferase located in the endoplasmic reticulum (ER), is required for lipidation of Wnt proteins to enable their trafficking from the ER in mammalian cell culture. However, it is unclear whether Porcn is required for trafficking of all members of the Wnt family. In this study, we investigated the function of Porcn in zebrafish embryos. We identified two zebrafish homologs of porcupine, porcn and porcupine-like (porcn-l). Zebrafish porcn, but not porcn-l, restores secretion of Wnt proteins in porcn-deficient mouse L cells. Morpholino-mediated knockdown of porcn in zebrafish embryos impairs convergence and extension (CE) during gastrulation without changing embryonic patterning. Moreover, porcn interacts genetically with wnt5b and wnt11 in regulating CE. By contrast, porcn-deficient embryos do not exhibit phenotypes caused by failure in canonical Wnt signaling, which is activated by several Wnt ligands, including Wnt3a. Furthermore, expression of genes regulated by the canonical Wnt signaling pathway is not perturbed in knockdown embryos relative to that in controls. Although the trafficking and lipidation of ectopically expressed zebrafish Wnt5b and mouse Wnt5a are impaired in porcn-deficient embryos, those of ectopically expressed Wnt3a are less or not affected. In addition, the secretion of Wnt5a is inhibited by less Porcn inhibitor than that of Wnt3a in HEK293T cells. Thus, a decrease of Porcn activity does not equivalently affect trafficking and lipidation of different Wnt proteins in zebrafish embryos and in cultured mammalian cells.
Collapse
Affiliation(s)
- Qiuhong Chen
- Okazaki Institute for Integrative Bioscience and National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | | | | |
Collapse
|
35
|
Miyake A, Nihno S, Murakoshi Y, Satsuka A, Nakayama Y, Itoh N. Neucrin, a novel secreted antagonist of canonical Wnt signaling, plays roles in developing neural tissues in zebrafish. Mech Dev 2012; 128:577-90. [PMID: 22265871 DOI: 10.1016/j.mod.2012.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 12/21/2011] [Accepted: 01/05/2012] [Indexed: 02/06/2023]
Abstract
Wnt signaling plays crucial roles in neural development. We previously identified Neucrin, a neural-specific secreted antagonist of canonical Wnt/β-catenin signaling, in humans and mice. Neucrin has one cysteine-rich domain, in which the positions of 10 cysteine residues are similar to those in the second cysteine-rich domain of Dickkopfs, secreted Wnt antagonists. Here, we have identified zebrafish neucrin to understand its roles in vivo. Zebrafish Neucrin also has one cysteine-rich domain, which is significantly similar to that of mouse Neucrin. Zebrafish neucrin was also predominantly expressed in developing neural tissues. To examine roles of neucrin in neural development, we analyzed neucrin knockdown embryos. Neural development in zebrafish embryos was impaired by the knockdown of neucrin. The knockdown of neucrin caused increased expression of the Wnt/β-catenin target genes. In contrast, overexpression of neucrin reduced the expression of the Wnt/β-catenin target genes. The knockdown of neucrin affected specification of dorsal region in the midbrain and hindbrain. The knockdown of neucrin also suppressed neuronal differentiation and caused increased cell proliferation and apoptosis in developing neural tissues. Neucrin is a unique secreted Wnt antagonist that is predominantly expressed in developing neural tissues and plays roles in neural development in zebrafish.
Collapse
Affiliation(s)
- Ayumi Miyake
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto 606-8501, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Sanchez-Simon F, Ledo A, Arevalo R, Rodriguez R. New insights into opioid regulatory pathways: influence of opioids on Wnt1 expression in zebrafish embryos. Neuroscience 2012; 200:237-47. [DOI: 10.1016/j.neuroscience.2011.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/14/2011] [Accepted: 10/15/2011] [Indexed: 01/24/2023]
|
37
|
Xin N, Benchabane H, Tian A, Nguyen K, Klofas L, Ahmed Y. Erect Wing facilitates context-dependent Wnt/Wingless signaling by recruiting the cell-specific Armadillo-TCF adaptor Earthbound to chromatin. Development 2011; 138:4955-67. [PMID: 22028028 DOI: 10.1242/dev.068890] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During metazoan development, the Wnt/Wingless signal transduction pathway is activated repetitively to direct cell proliferation, fate specification, differentiation and apoptosis. Distinct outcomes are elicited by Wnt stimulation in different cellular contexts; however, mechanisms that confer context specificity to Wnt signaling responses remain largely unknown. Starting with an unbiased forward genetic screen in Drosophila, we recently uncovered a novel mechanism by which the cell-specific co-factor Earthbound 1 (Ebd1), and its human homolog jerky, promote interaction between the Wnt pathway transcriptional co-activators β-catenin/Armadillo and TCF to facilitate context-dependent Wnt signaling responses. Here, through the same genetic screen, we find an unanticipated requirement for Erect Wing (Ewg), the fly homolog of the human sequence-specific DNA-binding transcriptional activator nuclear respiratory factor 1 (NRF1), in promoting contextual regulation of Wingless signaling. Ewg and Ebd1 functionally interact with the Armadillo-TCF complex and mediate the same context-dependent Wingless signaling responses. In addition, Ewg and Ebd1 have similar cell-specific expression profiles, bind to each other directly and also associate with chromatin at shared genomic sites. Furthermore, recruitment of Ebd1 to chromatin is abolished in the absence of Ewg. Our findings provide in vivo evidence that recruitment of a cell-specific co-factor complex to specific chromatin sites, coupled with its ability to facilitate Armadillo-TCF interaction and transcriptional activity, promotes contextual regulation of Wnt/Wingless signaling responses.
Collapse
Affiliation(s)
- Nan Xin
- Department of Genetics and Norris Cotton Cancer Center, Dartmouth Medical School, Hanover, NH 03755, USA
| | | | | | | | | | | |
Collapse
|
38
|
Weisinger K, Kohl A, Kayam G, Monsonego-Ornan E, Sela-Donenfeld D. Expression of hindbrain boundary markers is regulated by FGF3. Biol Open 2011; 1:67-74. [PMID: 23213398 PMCID: PMC3507201 DOI: 10.1242/bio.2011032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Compartment boundaries act as organizing centers that segregate adjacent areas into domains of gene expression and regulation, and control their distinct fates via the secretion of signalling factors. During hindbrain development, a specialized cell-population forms boundaries between rhombomeres. These boundary cells demonstrate unique morphological properties and express multiple genes that differs them from intra-rhombomeric cells. Yet, little is known regarding the mechanisms that controls the expression or function of these boundary markers.Multiple components of the FGF signaling system, including ligands, receptors, downstream effectors as well as proteoglycans are shown to localize to boundary cells in the chick hindbrain. These patterns raise the possibility that FGF signaling plays a role in regulating boundary properties. We provide evidence to the role of FGF signaling, particularly the boundary-derived FGF3, in regulating the expression of multiple markers at hindbrain boundaries. These findings enable further characterization of the unique boundary-cell population, and expose a new function for FGFs as regulators of boundary-gene expression in the chick hindbrain.
Collapse
|
39
|
Wullimann MF, Mueller T, Distel M, Babaryka A, Grothe B, Köster RW. The long adventurous journey of rhombic lip cells in jawed vertebrates: a comparative developmental analysis. Front Neuroanat 2011; 5:27. [PMID: 21559349 PMCID: PMC3085262 DOI: 10.3389/fnana.2011.00027] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 04/06/2011] [Indexed: 12/21/2022] Open
Abstract
This review summarizes vertebrate rhombic lip and early cerebellar development covering classic approaches up to modern developmental genetics which identifies the relevant differential gene expression domains and their progeny. Most of this information is derived from amniotes. However, progress in anamniotes, particularly in the zebrafish, has recently been made. The current picture suggests that rhombic lip and cerebellar development in jawed vertebrates (gnathostomes) share many characteristics. Regarding cerebellar development, these include a ptf1a expressing ventral cerebellar proliferation (VCP) giving rise to Purkinje cells and other inhibitory cerebellar cell types, and an atoh1 expressing upper rhombic lip giving rise to an external granular layer (EGL, i.e., excitatory granule cells) and an early ventral migration into the anterior rhombencephalon (cholinergic nuclei). As for the lower rhombic lip (LRL), gnathostome commonalities likely include the formation of precerebellar nuclei (mossy fiber origins) and partially primary auditory nuclei (likely convergently evolved) from the atoh1 expressing dorsal zone. The fate of the ptf1a expressing ventral LRL zone which gives rise to (excitatory cells of) the inferior olive (climbing fiber origin) and (inhibitory cells of ) cochlear nuclei in amniotes, has not been determined in anamniotes. Special for the zebrafish in comparison to amniotes is the predominant origin of anamniote excitatory deep cerebellar nuclei homologs (i.e., eurydendroid cells) from ptf1a expressing VCP cells, the sequential activity of various atoh1 paralogs and the incomplete coverage of the subpial cerebellar plate with proliferative EGL cells. Nevertheless, the conclusion that a rhombic lip and its major derivatives evolved with gnathostome vertebrates only and are thus not an ancestral craniate character complex is supported by the absence of a cerebellum (and likely absence of its afferent and efferent nuclei) in jawless fishes
Collapse
Affiliation(s)
- Mario F Wullimann
- Graduate School of Systemic Neurosciences and Department Biology II, Ludwig-Maximilians-Universität Munich Planegg, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Cox AA, Jezewski PA, Fang PK, Payne-Ferreira TL. Zebrafish Wnt9a,9b paralog comparisons suggest ancestral roles for Wnt9 in neural, oral-pharyngeal ectoderm and mesendoderm. Gene Expr Patterns 2010; 10:251-8. [PMID: 20566374 DOI: 10.1016/j.gep.2010.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 05/13/2010] [Accepted: 05/15/2010] [Indexed: 10/19/2022]
Abstract
The Wnts are a highly conserved family of secreted glycoproteins involved in cell-cell signaling and pattern formation during early embryonic development. Teasing out the role of individual Wnt molecules through development is challenging. Gene duplications are one of the most important mechanisms for generating evolutionary variations. The current consensus suggests that most anatomical variation is generated by divergence of regulatory control regions rather than by coding sequence divergence. Thus phylogenetic comparisons of divergent gene expression patterns are essential to understanding ancestral morphogenetic patterns from which subsequent anatomy diversified in modern lineages. We previously demonstrated strongest expression of zebrafish wnt9b within its heart tube, limb bud and ventral/anterior ectoderm during oral and pharyngeal arch patterning. Our goal is to compare and contrast zwnt9b to its closest paralog, zwnt9a. Sequenced, fulllength zebrafish wnt9a and wnt9b cDNA clones were used for phylogenetic analysis, which suggests their derivation from a common pre-vertebrate archeolog by gene duplication and divergence. Here we demonstrate that zwnt9a expression is found within unique (CNS, pronephric ducts, sensory organs) and overlapping (pectoral fin buds) expression domains relative to zwnt9b. Apparently, Wnt9 paralogs differentially parsed common ancestral expression domains during their subsequent rounds of gene duplication, divergence and loss in different vertebrate lineages. This expression data suggests ancestral roles for Wnt9s in early patterning of neural/oral-pharyngeal ectoderm and mesendoderm derivatives.
Collapse
Affiliation(s)
- A A Cox
- Department of Biology, University of Massachusetts, North Dartmouth, MA 02747, USA
| | | | | | | |
Collapse
|
41
|
Chordate roots of the vertebrate nervous system: expanding the molecular toolkit. Nat Rev Neurosci 2009; 10:736-46. [PMID: 19738625 DOI: 10.1038/nrn2703] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vertebrate brain is highly complex with millions to billions of neurons. During development, the neural plate border region gives rise to the neural crest, cranial placodes and, in anamniotes, to Rohon-Beard sensory neurons, whereas the boundary region of the midbrain and hindbrain develops organizer properties. Comparisons of developmental gene expression and neuroanatomy between vertebrates and the basal chordate amphioxus, which has only thousands of neurons and lacks a neural crest, most placodes and a midbrain-hindbrain organizer, indicate that these vertebrate features were built on a foundation already present in the ancestral chordate. Recent advances in genomics have provided insights into the elaboration of the molecular toolkit at the invertebrate-vertebrate transition that may have facilitated the evolution of these vertebrate characteristics.
Collapse
|
42
|
Paridaen JTML, Danesin C, Elas AT, van de Water S, Houart C, Zivkovic D. Apc1 is required for maintenance of local brain organizers and dorsal midbrain survival. Dev Biol 2009; 331:101-12. [PMID: 19397905 DOI: 10.1016/j.ydbio.2009.04.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/19/2009] [Accepted: 04/20/2009] [Indexed: 11/22/2022]
Abstract
The tumor suppressor Apc1 is an intracellular antagonist of the Wnt/beta-catenin pathway, which is vital for induction and patterning of the early vertebrate brain. However, its role in later brain development is less clear. Here, we examined the mechanisms underlying effects of an Apc1 zygotic-effect mutation on late brain development in zebrafish. Apc1 is required for maintenance of established brain subdivisions and control of local organizers such as the isthmic organizer (IsO). Caudal expansion of Fgf8 from IsO into the cerebellum is accompanied by hyperproliferation and abnormal cerebellar morphogenesis. Loss of apc1 results in reduced proliferation and apoptosis in the dorsal midbrain. Mosaic analysis shows that Apc is required cell-autonomously for maintenance of dorsal midbrain cell fate. The tectal phenotype occurs independently of Fgf8-mediated IsO function and is predominantly caused by stabilization of beta-catenin and subsequent hyperactivation of Wnt/beta-catenin signalling, which is mainly mediated through LEF1 activity. Chemical activation of the Wnt/beta-catenin in wild-type embryos during late brain maintenance stages phenocopies the IsO and tectal phenotypes of the apc mutants. These data demonstrate that Apc1-mediated restriction of Wnt/beta-catenin signalling is required for maintenance of local organizers and tectal integrity.
Collapse
|
43
|
Abstract
Wnt signaling regulates embryonic patterning and controls stem cell homeostasis, while aberrant Wnt activity is associated with disease. One Wnt family member, Wnt3, is required in mouse for specification of mesoderm, and later regulates neural patterning, apical ectodermal ridge formation, and hair growth. We have identified and performed preliminary characterization of the zebrafish wnt3 gene. wnt3 is expressed in the developing tailbud and neural tissue including the zona limitans intrathalamica (ZLI), optic tectum, midbrain-hindbrain boundary, and dorsal hindbrain and spinal cord. Expression in these regions suggests that Wnt3 participates in processes such as forebrain compartmentalization and regulation of tectal wiring topography by retinal ganglia axons. Surprisingly, wnt3 expression is not detectable during mesoderm specification, making it unlikely that Wnt3 regulates this process in zebrafish. This lack of early expression should make it possible to study later Wnt3-regulated patterning events, such as neural patterning, by knockdown studies in zebrafish.
Collapse
Affiliation(s)
- Wilson K. Clements
- Section of Cell and Developmental Biology, Division of Biological Sciences University of California at San Diego, La Jolla, CA 92093-0380
| | - Karen G. Ong
- Section of Cell and Developmental Biology, Division of Biological Sciences University of California at San Diego, La Jolla, CA 92093-0380
| | - David Traver
- Section of Cell and Developmental Biology, Division of Biological Sciences University of California at San Diego, La Jolla, CA 92093-0380
| |
Collapse
|
44
|
Expression of Wnt gene family and frizzled receptors in head and neck squamous cell carcinomas. Virchows Arch 2009; 455:67-75. [DOI: 10.1007/s00428-009-0793-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 04/09/2009] [Accepted: 05/17/2009] [Indexed: 11/28/2022]
|
45
|
Rhinn M, Lun K, Ahrendt R, Geffarth M, Brand M. Zebrafish gbx1 refines the midbrain-hindbrain boundary border and mediates the Wnt8 posteriorization signal. Neural Dev 2009; 4:12. [PMID: 19341460 PMCID: PMC2674439 DOI: 10.1186/1749-8104-4-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 04/02/2009] [Indexed: 12/27/2022] Open
Abstract
Background Studies in mouse, Xenopus and chicken have shown that Otx2 and Gbx2 expression domains are fundamental for positioning the midbrain-hindbrain boundary (MHB) organizer. Of the two zebrafish gbx genes, gbx1 is a likely candidate to participate in this event because its early expression is similar to that reported for Gbx2 in other species. Zebrafish gbx2, on the other hand, acts relatively late at the MHB. To investigate the function of zebrafish gbx1 within the early neural plate, we used a combination of gain- and loss-of-function experiments. Results We found that ectopic gbx1 expression in the anterior neural plate reduces forebrain and midbrain, represses otx2 expression and repositions the MHB to a more anterior position at the new gbx1/otx2 border. In the case of gbx1 loss-of-function, the initially robust otx2 domain shifts slightly posterior at a given stage (70% epiboly), as does MHB marker expression. We further found that ectopic juxtaposition of otx2 and gbx1 leads to ectopic activation of MHB markers fgf8, pax2.1 and eng2. This indicates that, in zebrafish, an interaction between otx2 and gbx1 determines the site of MHB development. Our work also highlights a novel requirement for gbx1 in hindbrain development. Using cell-tracing experiments, gbx1 was found to cell-autonomously transform anterior neural tissue into posterior. Previous studies have shown that gbx1 is a target of Wnt8 graded activity in the early neural plate. Consistent with this, we show that gbx1 can partially restore hindbrain patterning in cases of Wnt8 loss-of-function. We propose that in addition to its role at the MHB, gbx1 acts at the transcriptional level to mediate Wnt8 posteriorizing signals that pattern the developing hindbrain. Conclusion Our results provide evidence that zebrafish gbx1 is involved in positioning the MHB in the early neural plate by refining the otx2 expression domain. In addition to its role in MHB formation, we have shown that gbx1 is a novel mediator of Wnt8 signaling during hindbrain patterning.
Collapse
Affiliation(s)
- Muriel Rhinn
- Biotechnology Center, and Center for Regenerative Therapies Dresden, CRTD, Dresden University of Technology, Dresden, Germany.
| | | | | | | | | |
Collapse
|
46
|
Ninkovic J, Stigloher C, Lillesaar C, Bally-Cuif L. Gsk3beta/PKA and Gli1 regulate the maintenance of neural progenitors at the midbrain-hindbrain boundary in concert with E(Spl) factor activity. Development 2008; 135:3137-48. [PMID: 18725518 DOI: 10.1242/dev.020479] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal production in the midbrain-hindbrain domain (MH) of the vertebrate embryonic neural tube depends on a progenitor pool called the ;intervening zone' (IZ), located at the midbrain-hindbrain boundary. The progressive recruitment of IZ progenitors along the mediolateral (future dorsoventral) axis prefigures the earlier maturation of the MH basal plate. It also correlates with a lower sensitivity of medial versus lateral IZ progenitors to the neurogenesis inhibition process that maintains the IZ pool. This role is performed in zebrafish by the E(Spl) factors Her5 and Her11, but the molecular cascades cooperating with Her5/11, and those accounting for their reduced effect in the medial IZ, remain unknown. We demonstrate here that the kinases Gsk3beta and cAMP-dependent protein kinase A (PKA) are novel determinants of IZ formation and cooperate with E(Spl) activity in a dose-dependent manner. Similar to E(Spl), we show that the activity of Gsk3beta/PKA is sensed differently by medial versus lateral IZ progenitors. Furthermore, we identify the transcription factor Gli1, expressed in medial IZ cells, as an antagonist of E(Spl) and Gsk3beta/PKA, and demonstrate that the neurogenesis-promoting activity of Gli1 accounts for the reduced sensitivity of medial IZ progenitors to neurogenesis inhibitors and their increased propensity to differentiate. We also show that the expression and activity of Gli1 in this process are, surprisingly, independent of Hedgehog signaling. Together, our results suggest a model in which the modulation of E(Spl) and Gsk3beta/PKA activities by Gli1 underlies the dynamic properties of IZ maintenance and recruitment.
Collapse
Affiliation(s)
- Jovica Ninkovic
- Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Department of Zebrafish Neurogenetics, Institute of Developmental Genetics, Ingolstaedter Landstrasse 1, D-85764 Neuherberg, Germany
| | | | | | | |
Collapse
|
47
|
Wang L, Wang J, Wu Y, Wu J, Pang S, Pan R, Wen T. A novel function of dcf1 during the differentiation of neural stem cells in vitro. Cell Mol Neurobiol 2008; 28:887-94. [PMID: 18365309 PMCID: PMC11515049 DOI: 10.1007/s10571-008-9266-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 02/02/2008] [Indexed: 12/01/2022]
Abstract
The study of neural dendrite formation is of great significance both in theory and applications. However, the molecular mechanisms of regulation remain unclear. We previously described a novel EST, which has high homology with dentritic cell factors (DCF1), expressed differentially between undifferentiated and differentiated neural stem cells (NSCs). In this study, we cloned, expressed, and silenced the dcf1 gene and offered insight into its function in regulating dendrite formation during the differentiation of NSCs. The results indicated that dcf1 encoded a 42 kD protein and could be successfully expressed both in Escherichia coli and NSCs. In order to silence dcf1 gene, three different kinds of siRNA vectors were constructed and transformed into the NSC line C17.2 and primary NSCs, resulting in down regulation of the dcf1 mRNA. Analysis of immunofluorescence or GFP illuminated that with overexpression of the dcf1 gene, the NSCs were maintained in undifferentiated status. After the dcf1 gene was silenced, cells tended to differentiate into neurons and astrocytes.
Collapse
Affiliation(s)
- Lei Wang
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Jiao Wang
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
- Institute of systems biology, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Yiliu Wu
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
- Institute of systems biology, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Jie Wu
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Shuya Pang
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Rong Pan
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| | - Tieqiao Wen
- The Laboratory of neural molecular biology, School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
- Institute of systems biology, Shanghai University, 99 Shangda Road, Shanghai, 200444 China
| |
Collapse
|
48
|
Abstract
Wnt signaling plays several important roles in the development of the zebrafish central nervous system (CNS). This chapter outlines both the known and postulated roles of Wnts from the earliest step of neural plate induction to relatively late events such as axon pathfinding and synaptogenesis. The common tools useful for examining Wnt function and nervous system development in zebrafish are first reviewed. Examples are then provided for specific phenotypes resulting from gain and loss of Wnt activity at multiple developmental stages. Finally, specific assays and reagents that can be used to investigate the function of novel Wnt pathway components in CNS development are listed.
Collapse
Affiliation(s)
- Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
49
|
Verkade H, Heath JK. Wnt signaling mediates diverse developmental processes in zebrafish. Methods Mol Biol 2008; 469:225-51. [PMID: 19109714 DOI: 10.1007/978-1-60327-469-2_17] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A combination of forward and reverse genetic approaches in zebrafish has revealed novel roles for canonical Wnt and Wnt/PCP signaling during vertebrate development. Forward genetics in zebrafish provides an exceptionally powerful tool to assign roles in vertebrate developmental processes to novel genes, as well as elucidating novel roles played by known genes. This has indeed turned out to be the case for components of the canonical Wnt signaling pathway. Non-canonical Wnt signaling in the zebrafish is also currently a topic of great interest, due to the identified roles of this pathway in processes requiring the integration of cell polarity and cell movement, such as the directed migration movements that drive the narrowing and lengthening (convergence and extension) of the embryo during early development.
Collapse
Affiliation(s)
- Heather Verkade
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
50
|
Hogan BM, Verkade H, Lieschke GJ, Heath JK. Manipulation of gene expression during zebrafish embryonic development using transient approaches. Methods Mol Biol 2008; 469:273-300. [PMID: 19109716 DOI: 10.1007/978-1-60327-469-2_19] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The rapid embryonic development and high fecundity of zebrafish contribute to the great advantages of this model for the study of developmental genetics. Transient disruption of the normal function of a gene during development can be achieved by microinjecting mRNA, DNA or short chemically stabilized anti-sense oligomers, called morpholinos (MOs), into early zebrafish embryos. The ensuing develop ment of the microinjected embryos is observed over the following hours and days to analyze the impact of the microinjected products on embryogenesis. Compared to stable reverse genetic approaches (sta ble transgenesis, targeted mutants recovered by TILLING), these transient reverse genetic approaches are vastly quicker, relatively affordable, and require little animal facility space. Common applications of these methodologies allow analysis of gain-of-function (gene overexpression or dominant active), loss-of-function (gene knock down or dominant negative), mosaic analysis, lineage-restricted studies and cell tracing experiments. The use of these transient approaches for the manipulation of gene expression has improved our understanding of many key developmental pathways including both the Wnt/beta-catenin and Wnt/PCP pathways, as covered in some detail in Chapter 17 of this book. This chapter describes the most common and versatile approaches: gain of function and loss of function using DNA and mRNA injections and loss of function using MOs.
Collapse
Affiliation(s)
- Benjamin M Hogan
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584, CT Utrecht, The Netherlands
| | | | | | | |
Collapse
|