1
|
Hidalgo-Sánchez M, Sánchez-Guardado L, Rodríguez-León J, Francisco-Morcillo J. The role of FGF15/FGF19 in the development of the central nervous system, eyes and inner ears in vertebrates. Tissue Cell 2024; 91:102619. [PMID: 39579736 DOI: 10.1016/j.tice.2024.102619] [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: 07/23/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Fibroblast growth factor 19 (FGF19), and its rodent ortholog FGF15, is a member of a FGF subfamily directly involved in metabolism, acting in an endocrine way. During embryonic development, FGF15/FGF19 also functions as a paracrine or autocrine factor, regulating key events in a large number of organs. In this sense, the Fgf15/Fgf19 genes control the correct development of the brain, eye, inner ear, heart, pharyngeal pouches, tail bud and limbs, among other organs, as well as muscle growth in adulthood. These growth factors show relevant differences according to molecular structures, signalling pathway and function. Moreover, their expression patterns are highly dynamic at different stages of development, in particular in the central nervous system. The difficulty in understanding the action of these genes increases when comparing their expression patterns and regulatory mechanisms between different groups of vertebrates. The present review will address the expression patterns and functions of the Fgf15/Fgf19 genes at different stages of vertebrate embryonic development, with special attention to the regulation of the early specification, cell differentiation, and morphogenesis of the central nervous system and some sensory organs such as eye and inner ear. The most relevant anatomical aspects related to the structures analysed have also been considered in detail to provide an understandable context for the molecular and cellular studies shown.
Collapse
Affiliation(s)
- Matías Hidalgo-Sánchez
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain.
| | - Luis Sánchez-Guardado
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía Humana, Facultad de Medicina y Ciencias de la Salud, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| |
Collapse
|
2
|
Kim CN, Shin D, Wang A, Nowakowski TJ. Spatiotemporal molecular dynamics of the developing human thalamus. Science 2023; 382:eadf9941. [PMID: 37824646 PMCID: PMC10758299 DOI: 10.1126/science.adf9941] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/15/2023] [Indexed: 10/14/2023]
Abstract
The thalamus plays a central coordinating role in the brain. Thalamic neurons are organized into spatially distinct nuclei, but the molecular architecture of thalamic development is poorly understood, especially in humans. To begin to delineate the molecular trajectories of cell fate specification and organization in the developing human thalamus, we used single-cell and multiplexed spatial transcriptomics. We show that molecularly defined thalamic neurons differentiate in the second trimester of human development and that these neurons organize into spatially and molecularly distinct nuclei. We identified major subtypes of glutamatergic neuron subtypes that are differentially enriched in anatomically distinct nuclei and six subtypes of γ-aminobutyric acid-mediated (GABAergic) neurons that are shared and distinct across thalamic nuclei.
Collapse
Affiliation(s)
- Chang N Kim
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - David Shin
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
3
|
Kim CN, Shin D, Wang A, Nowakowski TJ. Spatiotemporal molecular dynamics of the developing human thalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554174. [PMID: 37662287 PMCID: PMC10473600 DOI: 10.1101/2023.08.21.554174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The thalamus plays a central coordinating role in the brain. Thalamic neurons are organized into spatially-distinct nuclei, but the molecular architecture of thalamic development is poorly understood, especially in humans. To begin to delineate the molecular trajectories of cell fate specification and organization in the developing human thalamus, we used single cell and multiplexed spatial transcriptomics. Here we show that molecularly-defined thalamic neurons differentiate in the second trimester of human development, and that these neurons organize into spatially and molecularly distinct nuclei. We identify major subtypes of glutamatergic neuron subtypes that are differentially enriched in anatomically distinct nuclei. In addition, we identify six subtypes of GABAergic neurons that are shared and distinct across thalamic nuclei. One-Sentence Summary Single cell and spatial profiling of the developing thalamus in the first and second trimester yields molecular mechanisms of thalamic nuclei development.
Collapse
|
4
|
Sampath Kumar A, Tian L, Bolondi A, Hernández AA, Stickels R, Kretzmer H, Murray E, Wittler L, Walther M, Barakat G, Haut L, Elkabetz Y, Macosko EZ, Guignard L, Chen F, Meissner A. Spatiotemporal transcriptomic maps of whole mouse embryos at the onset of organogenesis. Nat Genet 2023; 55:1176-1185. [PMID: 37414952 PMCID: PMC10335937 DOI: 10.1038/s41588-023-01435-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/25/2023] [Indexed: 07/08/2023]
Abstract
Spatiotemporal orchestration of gene expression is required for proper embryonic development. The use of single-cell technologies has begun to provide improved resolution of early regulatory dynamics, including detailed molecular definitions of most cell states during mouse embryogenesis. Here we used Slide-seq to build spatial transcriptomic maps of complete embryonic day (E) 8.5 and E9.0, and partial E9.5 embryos. To support their utility, we developed sc3D, a tool for reconstructing and exploring three-dimensional 'virtual embryos', which enables the quantitative investigation of regionalized gene expression patterns. Our measurements along the main embryonic axes of the developing neural tube revealed several previously unannotated genes with distinct spatial patterns. We also characterized the conflicting transcriptional identity of 'ectopic' neural tubes that emerge in Tbx6 mutant embryos. Taken together, we present an experimental and computational framework for the spatiotemporal investigation of whole embryonic structures and mutant phenotypes.
Collapse
Affiliation(s)
- Abhishek Sampath Kumar
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Luyi Tian
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adriano Bolondi
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Amèlia Aragonés Hernández
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Robert Stickels
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lars Wittler
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maria Walther
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Gabriel Barakat
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Leah Haut
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Yechiel Elkabetz
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Evan Z Macosko
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Léo Guignard
- Aix Marseille University, Toulon University, Centre National de la Recherche Scientifique, Laboratoire d'Informatique et Systèmes 7020, Turing Centre for Living Systems, Marseille, France
| | - Fei Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
5
|
Sun M, Zhou D, Wu J, Zhou J, Xu J. Sdy-1 Executes Antitumor Activity in HepG2 and HeLa Cancer Cells by Inhibiting the Wnt/β-Catenin Signaling Pathway. Mar Drugs 2022; 20:md20020125. [PMID: 35200654 PMCID: PMC8877534 DOI: 10.3390/md20020125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/24/2022] Open
Abstract
Demethylincisterol A3 (Sdy-1), a highly degraded sterol that we previously isolated from Chinese mangrove Rhizophora mucronata endophytic Pestalotiopsis sp. HQD-6, exhibits potent antitumor activity towards a variety of cancer cells. In this study, we further verified that Sdy-1 effectively inhibited the proliferation and migration of human liver (HepG2) and cervical cancer (HeLa) cells in vitro and it can induce cell apoptosis and arrest the cell cycle in the G1-phase. Mechanistically, we demonstrated that Sdy-1 executes its function via inhibition of the Wnt/β-catenin signaling pathway. Sdy-1 may not inhibit the Wnt signaling pathway through the cascade reaction from upstream to downstream, but directly acts on β-catenin to reduce its transcription level, thereby reducing the level of β-catenin protein and further reducing the expression of downstream related proteins. The possible interaction between Sdy-1 and β-catenin protein was further confirmed by molecular docking studies. In the nude mouse xenograft model, Sdy-1 can also significantly inhibit tumor growth. These results indicated that Sdy-1 is an efficient inhibitor of the Wnt signaling pathway and is a promising antitumor candidate for therapeutic applications.
Collapse
Affiliation(s)
- Mengyu Sun
- One Health Institute, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, China; (M.S.); (D.Z.); (J.W.)
| | - Dongdong Zhou
- One Health Institute, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, China; (M.S.); (D.Z.); (J.W.)
| | - Jingwan Wu
- One Health Institute, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, China; (M.S.); (D.Z.); (J.W.)
| | - Jing Zhou
- Hainan Provincial Fine Chemical Engineering Research Center, School of Life Sciences, Hainan University, Haikou 570228, China;
| | - Jing Xu
- One Health Institute, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, China; (M.S.); (D.Z.); (J.W.)
- Correspondence:
| |
Collapse
|
6
|
Company V, Moreno-Cerdá A, Andreu-Cervera A, Murcia-Ramón R, Almagro-García F, Echevarría D, Martínez S, Puelles E. Wnt1 Role in the Development of the Habenula and the Fasciculus Retroflexus. Front Cell Dev Biol 2021; 9:755729. [PMID: 34722541 PMCID: PMC8551717 DOI: 10.3389/fcell.2021.755729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022] Open
Abstract
Wnt1 is one of the morphogenes that controls the specification and differentiation of neuronal populations in the developing central nervous system. The habenula is a diencephalic neuronal complex located in the most dorsal aspect of the thalamic prosomere. This diencephalic neuronal population is involved in the limbic system and its malfunction is related with several psychiatric disorders. Our aim is to elucidate the Wnt1 role in the habenula and its main efferent tract, the fasciculus retroflexus, development. In order to achieve these objectives, we analyzed these structures development in a Wnt1 lack of function mouse model. The habenula was generated in our model, but it presented an enlarged volume. This alteration was due to an increment in habenular neuroblasts proliferation rate. The fasciculus retroflexus also presented a wider and disorganized distribution and a disturbed final trajectory toward its target. The mid-hindbrain territories that the tract must cross were miss-differentiated in our model. The specification of the habenula is Wnt1 independent. Nevertheless, it controls its precursors proliferation rate. Wnt1 expressed in the isthmic organizer is vital to induce the midbrain and rostral hindbrain territories. The alteration of these areas is responsible for the fasciculus retroflexus axons misroute.
Collapse
Affiliation(s)
- Verónica Company
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Ana Moreno-Cerdá
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Abraham Andreu-Cervera
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Raquel Murcia-Ramón
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Francisca Almagro-García
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Diego Echevarría
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Salvador Martínez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - Eduardo Puelles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| |
Collapse
|
7
|
Sugahara F, Murakami Y, Pascual-Anaya J, Kuratani S. Forebrain Architecture and Development in Cyclostomes, with Reference to the Early Morphology and Evolution of the Vertebrate Head. BRAIN, BEHAVIOR AND EVOLUTION 2021; 96:305-317. [PMID: 34537767 DOI: 10.1159/000519026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022]
Abstract
The vertebrate head and brain are characterized by highly complex morphological patterns. The forebrain, the most anterior division of the brain, is subdivided into the diencephalon, hypothalamus, and telencephalon from the neuromeric subdivision into prosomeres. Importantly, the telencephalon contains the cerebral cortex, which plays a key role in higher order cognitive functions in humans. To elucidate the evolution of the forebrain regionalization, comparative analyses of the brain development between extant jawed and jawless vertebrates are crucial. Cyclostomes - lampreys and hagfishes - are the only extant jawless vertebrates, and diverged from jawed vertebrates (gnathostomes) over 500 million years ago. Previous developmental studies on the cyclostome brain were conducted mainly in lampreys because hagfish embryos were rarely available. Although still scarce, the recent availability of hagfish embryos has propelled comparative studies of brain development and gene expression. By integrating findings with those of cyclostomes and fossil jawless vertebrates, we can depict the morphology, developmental mechanism, and even the evolutionary path of the brain of the last common ancestor of vertebrates. In this review, we summarize the development of the forebrain in cyclostomes and suggest what evolutionary changes each cyclostome lineage underwent during brain evolution. In addition, together with recent advances in the head morphology in fossil vertebrates revealed by CT scanning technology, we discuss how the evolution of craniofacial morphology and the changes of the developmental mechanism of the forebrain towards crown gnathostomes are causally related.
Collapse
Affiliation(s)
- Fumiaki Sugahara
- Division of Biology, Hyogo College of Medicine, Nishinomiya, Japan.,Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering, Ehime University, Matsuyama, Japan
| | - Juan Pascual-Anaya
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan.,Department of Animal Biology, Faculty of Science, University of Málaga, Málaga, Spain.,Andalusian Centre for Nanomedicine and Biotechnology (BIONAND), Málaga, Spain
| | - Shigeru Kuratani
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan.,Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| |
Collapse
|
8
|
Cheng AH, Cheng HYM. Genesis of the Master Circadian Pacemaker in Mice. Front Neurosci 2021; 15:659974. [PMID: 33833665 PMCID: PMC8021851 DOI: 10.3389/fnins.2021.659974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Peng C, Lu Y, Ren X, Wang Y, Zhang S, Chen M, Liu J, Fang F, Li T, Han J. Comprehensive bioinformatic analysis of Wnt1 and Wnt1-associated diseases. Intractable Rare Dis Res 2020; 9:14-22. [PMID: 32201670 PMCID: PMC7062594 DOI: 10.5582/irdr.2020.01018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Wnt1 is the first member of the Wnt family that was identified. It is phylogenetically conserved and essential for oncogenesis and multiple developmental processes. This study has summarized diseases and mutations related to Wnt1. Wnt1 is involved in various cancers, genetic type XV osteogenesis imperfecta, osteoporosis, and neurological diseases. The expression of Wnt1 in normal tissues and different types of cancers and the potential survival of cancer were analyzed using experiment-based bioinformatic analysis. Systematic analysis indicated that abnormal expression of Wnt1 is significantly associated with cancers, such as kidney renal carcinoma, hepatocellular carcinoma, thyroid carcinoma, head and neck squamous cell carcinoma, and uterine corpus endometrial carcinoma. GeneMANIA and STRING predicted that 32 proteins were involved with Wnt1 in Wnt signaling pathways and sorting and secretion of Wnts. These interacting molecules significantly co-occurred according to cBioPortal analysis. Thirty-three genes with an alteration frequency of more than 50% were observed in several cancers like esophageal squamous cell carcinoma, melanoma, and non-small cell lung cancer. Functional and experiment-based bioinformatics indicated that Wnt1 may act as a target of a potential biomarker for various types of human cancers. Wnt1 and other Wnt1-related proteins and signaling pathways may be ways to treat osteoporosis.
Collapse
Affiliation(s)
- Chuanming Peng
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Ji'nan, China
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China
| | - Yanqin Lu
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Ji'nan, China
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China
- Address correspondence to:Yanqin Lu and Jinxiang Han, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Ji'nan 250062, China. E-mail: ,
| | - Xiuzhi Ren
- Orthopaedic Surgery, The People's Hospital of Wuqing District, Tianjin, China
| | - Yanzhou Wang
- Shandong Provincial Hospital Affiliated with Shandong First Medical University, Ji'nan, China
| | - Shie Zhang
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Ji'nan, China
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China
| | - Mei Chen
- Orthopaedic Surgery, The People's Hospital of Wuqing District, Tianjin, China
| | - Junlong Liu
- Orthopaedic Surgery, The People's Hospital of Wuqing District, Tianjin, China
| | - Fengling Fang
- Orthopaedic Surgery, The People's Hospital of Wuqing District, Tianjin, China
| | - Tianyou Li
- Shandong Provincial Hospital Affiliated with Shandong First Medical University, Ji'nan, China
| | - Jinxiang Han
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Ji'nan, China
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, China
- Address correspondence to:Yanqin Lu and Jinxiang Han, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Ji'nan 250062, China. E-mail: ,
| |
Collapse
|
10
|
Tran H, Park W, Seong S, Jeong J, Nguyen Q, Yoon J, Baek K, Jeong Y. Tcf7l2
transcription factor is required for the maintenance, but not the initial specification, of the neurotransmitter identity in the caudal thalamus. Dev Dyn 2019; 249:646-655. [DOI: 10.1002/dvdy.146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/15/2019] [Accepted: 12/15/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Hong‐Nhung Tran
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Wonbae Park
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Sojeong Seong
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Ji‐eun Jeong
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Quy‐Hoai Nguyen
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Jaeseung Yoon
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Kwanghee Baek
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| | - Yongsu Jeong
- Department of Genetic Engineering, College of Life Sciences and Graduate School of BiotechnologyKyung Hee University Yongin‐si Republic of Korea
| |
Collapse
|
11
|
Collins JE, White RJ, Staudt N, Sealy IM, Packham I, Wali N, Tudor C, Mazzeo C, Green A, Siragher E, Ryder E, White JK, Papatheodoru I, Tang A, Füllgrabe A, Billis K, Geyer SH, Weninger WJ, Galli A, Hemberger M, Stemple DL, Robertson E, Smith JC, Mohun T, Adams DJ, Busch-Nentwich EM. Common and distinct transcriptional signatures of mammalian embryonic lethality. Nat Commun 2019; 10:2792. [PMID: 31243271 PMCID: PMC6594971 DOI: 10.1038/s41467-019-10642-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 05/22/2019] [Indexed: 12/20/2022] Open
Abstract
The Deciphering the Mechanisms of Developmental Disorders programme has analysed the morphological and molecular phenotypes of embryonic and perinatal lethal mouse mutant lines in order to investigate the causes of embryonic lethality. Here we show that individual whole-embryo RNA-seq of 73 mouse mutant lines (>1000 transcriptomes) identifies transcriptional events underlying embryonic lethality and associates previously uncharacterised genes with specific pathways and tissues. For example, our data suggest that Hmgxb3 is involved in DNA-damage repair and cell-cycle regulation. Further, we separate embryonic delay signatures from mutant line-specific transcriptional changes by developing a baseline mRNA expression catalogue of wild-type mice during early embryogenesis (4-36 somites). Analysis of transcription outside coding sequence identifies deregulation of repetitive elements in Morc2a mutants and a gene involved in gene-specific splicing. Collectively, this work provides a large scale resource to further our understanding of early embryonic developmental disorders.
Collapse
Affiliation(s)
- John E Collins
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Richard J White
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Nicole Staudt
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Ian M Sealy
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Ian Packham
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Neha Wali
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Cecilia Mazzeo
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Angela Green
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Emma Siragher
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Edward Ryder
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Jacqueline K White
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Irene Papatheodoru
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK
| | - Amy Tang
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK
| | - Anja Füllgrabe
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK
| | - Konstantinos Billis
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK
| | - Stefan H Geyer
- Division of Anatomy, MIC, Medical University of Vienna, Waehringerstr. 13, 1090, Wien, Austria
| | - Wolfgang J Weninger
- Division of Anatomy, MIC, Medical University of Vienna, Waehringerstr. 13, 1090, Wien, Austria
| | - Antonella Galli
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Myriam Hemberger
- The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
- Departments of Biochemistry & Molecular Biology and Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Derek L Stemple
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
- Camena Bioscience, The Science Village, Chesterford Research Park, Cambridge, CB10 1XL, UK
| | - Elizabeth Robertson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - James C Smith
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Timothy Mohun
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - David J Adams
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Elisabeth M Busch-Nentwich
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK.
| |
Collapse
|
12
|
Nakagawa Y. Development of the thalamus: From early patterning to regulation of cortical functions. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e345. [PMID: 31034163 DOI: 10.1002/wdev.345] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
The thalamus is a brain structure of the vertebrate diencephalon that plays a central role in regulating diverse functions of the cerebral cortex. In traditional view of vertebrate neuroanatomy, the thalamus includes three regions, dorsal thalamus, ventral thalamus, and epithalamus. Recent molecular embryological studies have redefined the thalamus and the associated axial nomenclature of the diencephalon in the context of forebrain patterning. This new view has provided a useful conceptual framework for studies on molecular mechanisms of patterning, neurogenesis and fate specification in the thalamus as well as the guidance mechanisms for thalamocortical axons. Additionally, the availability of genetic tools in mice has led to important findings on how thalamic development is linked to the development of other brain regions, particularly the cerebral cortex. This article will give an overview of the organization of the embryonic thalamus and how progenitor cells in the thalamus generate neurons that are organized into discrete nuclei. I will then discuss how thalamic development is orchestrated with the development of the cerebral cortex and other brain regions. This article is categorized under: Nervous System Development > Vertebrates: Regional Development Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Yasushi Nakagawa
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
13
|
Guo Q, Li JYH. Defining developmental diversification of diencephalon neurons through single cell gene expression profiling. Development 2019; 146:dev174284. [PMID: 30872278 PMCID: PMC6602344 DOI: 10.1242/dev.174284] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/07/2019] [Indexed: 12/31/2022]
Abstract
The embryonic diencephalon forms integration centers and relay stations in the forebrain. Anecdotal expression studies suggest that the diencephalon contains multiple developmental compartments and subdivisions. Here, we utilized single cell RNA sequencing to profile transcriptomes of dissociated cells from the diencephalon of E12.5 mouse embryos. We identified the divergence of different progenitors, intermediate progenitors, and emerging neurons. By mapping the identified cell groups to their spatial origins, we characterized the molecular features of cell types and cell states arising from various diencephalic domains. Furthermore, we reconstructed the developmental trajectory of distinct cell lineages, and thereby identified the genetic cascades and gene regulatory networks underlying the progression of the cell cycle, neurogenesis and cellular diversification. The analysis provides new insights into the molecular mechanisms underlying the amplification of intermediate progenitor cells in the thalamus. The single cell-resolved trajectories not only confirm a close relationship between the rostral thalamus and prethalamus, but also uncover an unexpected close relationship between the caudal thalamus, epithalamus and rostral pretectum. Our data provide a useful resource for systematic studies of cell heterogeneity and differentiation kinetics within the diencephalon.
Collapse
Affiliation(s)
- Qiuxia Guo
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - James Y H Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
- Institute for Systems Genomics, University of Connecticut, 400 Farmington Avenue, Farmington, CT 06030-6403, USA
| |
Collapse
|
14
|
The Ciliopathy Gene Ftm/Rpgrip1l Controls Mouse Forebrain Patterning via Region-Specific Modulation of Hedgehog/Gli Signaling. J Neurosci 2019; 39:2398-2415. [PMID: 30692221 PMCID: PMC6435827 DOI: 10.1523/jneurosci.2199-18.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/22/2018] [Accepted: 01/15/2019] [Indexed: 02/07/2023] Open
Abstract
Primary cilia are essential for CNS development. In the mouse, they play a critical role in patterning the spinal cord and telencephalon via the regulation of Hedgehog/Gli signaling. However, despite the frequent disruption of this signaling pathway in human forebrain malformations, the role of primary cilia in forebrain morphogenesis has been little investigated outside the telencephalon. Here we studied development of the diencephalon, hypothalamus and eyes in mutant mice in which the Ftm/Rpgrip1l ciliopathy gene is disrupted. At the end of gestation, Ftm−/− fetuses displayed anophthalmia, a reduction of the ventral hypothalamus and a disorganization of diencephalic nuclei and axonal tracts. In Ftm−/− embryos, we found that the ventral forebrain structures and the rostral thalamus were missing. Optic vesicles formed but lacked the optic cups. In Ftm−/− embryos, Sonic hedgehog (Shh) expression was virtually lost in the ventral forebrain but maintained in the zona limitans intrathalamica (ZLI), the mid-diencephalic organizer. Gli activity was severely downregulated but not lost in the ventral forebrain and in regions adjacent to the Shh-expressing ZLI. Reintroduction of the repressor form of Gli3 into the Ftm−/− background restored optic cup formation. Our data thus uncover a complex role of cilia in development of the diencephalon, hypothalamus and eyes via the region-specific control of the ratio of activator and repressor forms of the Gli transcription factors. They call for a closer examination of forebrain defects in severe ciliopathies and for a search for ciliopathy genes as modifiers in other human conditions with forebrain defects. SIGNIFICANCE STATEMENT The Hedgehog (Hh) signaling pathway is essential for proper forebrain development as illustrated by a human condition called holoprosencephaly. The Hh pathway relies on primary cilia, cellular organelles that receive and transduce extracellular signals and whose dysfunctions lead to rare inherited diseases called ciliopathies. To date, the role of cilia in the forebrain has been poorly studied outside the telencephalon. In this paper we study the role of the Ftm/Rpgrip1l ciliopathy gene in mouse forebrain development. We uncover complex functions of primary cilia in forebrain morphogenesis through region-specific modulation of the Hh pathway. Our data call for further examination of forebrain defects in ciliopathies and for a search for ciliopathy genes as modifiers in human conditions affecting forebrain development.
Collapse
|
15
|
Radial glia fibers translate Fgf8 morphogenetic signals to generate a thalamic nuclear complex protomap in the mantle layer. Brain Struct Funct 2018; 224:661-679. [PMID: 30470893 PMCID: PMC6420463 DOI: 10.1007/s00429-018-1794-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 11/09/2018] [Indexed: 01/25/2023]
Abstract
Thalamic neurons are distributed between different nuclear groups of the thalamic multinuclear complex; they develop topologically ordered specific projections that convey information on voluntary motor programs and sensory modalities to functional areas in the cerebral cortex. Since thalamic neurons present a homogeneous morphology, their functional specificity is derived from their afferent and efferent connectivity. Adequate development of thalamic afferent and efferent connections depends on guide signals that bind receptors in nuclear neuropils and axonal growth cones, respectively. These are finally regulated by regionalization processes in the thalamic neurons, codifying topological information. In this work, we studied the role of Fgf8 morphogenetic signaling in establishing the molecular thalamic protomap, which was revealed by Igsf21, Pde10a and Btbd3 gene expression in the thalamic mantle layer. Fgf8 signaling activity was evidenced by pERK expression in radial glia cells and fibers, which may represent a scaffold that translates neuroepithelial positional information to the mantle layer. In this work, we describe the fact that Fgf8-hypomorphic mice did not express pERK in radial glia cells and fibers and presented disorganized thalamic regionalization, increasing neuronal death in the ventro-lateral thalamus and strong disruption of thalamocortical projections. In conclusion, Fgf8 encodes the positional information required for thalamic nuclear regionalization and the development of thalamocortical projections.
Collapse
|
16
|
Newman EA, Wu D, Taketo MM, Zhang J, Blackshaw S. Canonical Wnt signaling regulates patterning, differentiation and nucleogenesis in mouse hypothalamus and prethalamus. Dev Biol 2018; 442:236-248. [PMID: 30063881 DOI: 10.1016/j.ydbio.2018.07.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023]
Abstract
The hypothalamus is a small, but anatomically and functionally complex region of the brain whose development is poorly understood. In this study, we have explored its development by studying the canonical Wnt signaling pathway, generating gain and loss of function mutations of beta-catenin (Ctnnb1) in both hypothalamic and prethalamic neuroepithelium. Deletion of Ctnnb1 resulted in an anteriorized and hypoplastic hypothalamus. Posterior structures were lost or reduced, and anterior structures were expanded. In contrast, overexpression of a constitutively active mutant form of Ctnnb1 resulted in severe hyperplasia of prethalamus and hypothalamus, and expanded expression of a subset of posterior and premamillary hypothalamic markers. Moderate defects in differentiation of Arx-positive GABAergic neural precursors were observed in both prethalamus and hypothalamus of Ctnnb1 loss of function mutants, while in gain of function mutants, their differentiation was completely suppressed, although markers of prethalamic progenitors were preserved. Multiple other region-specific markers, including several specific posterior hypothalamic structures, were also suppressed in Ctnnb1 gain of function mutations. Severe, region-specific defects in hypothalamic nucleogenesis were also observed in both gain and loss of function mutations of Ctnnb1. Finally, both gain and loss of function of Ctnnb1 also produced severe, non-cell autonomous disruptions of pituitary development. These findings demonstrate a central and multifaceted role for canonical Wnt signaling in regulating growth, patterning, differentiation and nucleogenesis in multiple diencephalic regions.
Collapse
Affiliation(s)
- Elizabeth A Newman
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan Wu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jiangyang Zhang
- Department of Radiology, NYU Langone School of Medicine, New York, NY, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
17
|
Cannonier SA, Gonzales CB, Ely K, Guelcher SA, Sterling JA. Hedgehog and TGFβ signaling converge on Gli2 to control bony invasion and bone destruction in oral squamous cell carcinoma. Oncotarget 2018; 7:76062-76075. [PMID: 27738315 PMCID: PMC5340177 DOI: 10.18632/oncotarget.12584] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the sixth most common cancer worldwide. OSCC invasion into the lymph nodes and mandible correlates with increased rates of recurrence and lower overall survival. Tumors that infiltrate mandibular bone proliferate rapidly and induce bone destruction. While survival rates have increased 12% over the last 20 years, this improvement is attributed to general advances in prevention, earlier detection, and updated treatments. Additionally, despite decades of research, the molecular mechanisms of OSCC invasion into the mandible are not well understood. Parathyroid Hormone-related Protein (PTHrP), has been shown to be essential for mandibular invasion in OSCC animal models, and our previous studies demonstrate that the transcription factor Gli2 increases PTHrP expression in tumor metastasis to bone. In OSCC, we investigated regulators of Gli2, including Hedgehog, TGFβ, and Wnt signaling to elucidate how PTHrP expression is controlled. Here we show that canonical Hedgehog and TGFβ signaling cooperate to increase PTHrP expression and mandibular invasion in a Gli2-dependent manner. Additionally, in an orthotopic model of mandibular invasion, inhibition of Gli2 using shRNA resulted in a significant decrease of both PTHrP expression and bony invasion. Collectively, our findings demonstrate that multiple signaling pathways converge on Gli2 to mediate PTHrP expression and bony invasion, highlighting Gli2 as a therapeutic target to prevent bony invasion in OSCC.
Collapse
Affiliation(s)
- Shellese A Cannonier
- Department of Veteran Affairs, Tennessee Valley Healthcare System, Nashville TN 37212, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville TN 37232, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville TN 37232, USA
| | - Cara B Gonzales
- Department of Comprehensive Dentistry, University of Texas Health Science Center San Antonio Dental School, San Antonio, TX 78229, USA
| | - Kim Ely
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville TN 37232, USA
| | - Scott A Guelcher
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville TN 37232, USA.,Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville TN 37235, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville TN 37235, USA
| | - Julie A Sterling
- Department of Veteran Affairs, Tennessee Valley Healthcare System, Nashville TN 37212, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville TN 37232, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville TN 37232, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville TN 37235, USA.,Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232, USA
| |
Collapse
|
18
|
Lee M, Yoon J, Song H, Lee B, Lam DT, Yoon J, Baek K, Clevers H, Jeong Y. Tcf7l2 plays crucial roles in forebrain development through regulation of thalamic and habenular neuron identity and connectivity. Dev Biol 2017; 424:62-76. [DOI: 10.1016/j.ydbio.2017.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 11/28/2022]
|
19
|
Diencephalic Size Is Restricted by a Novel Interplay Between GCN5 Acetyltransferase Activity and Retinoic Acid Signaling. J Neurosci 2017; 37:2565-2579. [PMID: 28154153 DOI: 10.1523/jneurosci.2121-16.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 01/20/2023] Open
Abstract
Diencephalic defects underlie an array of neurological diseases. Previous studies have suggested that retinoic acid (RA) signaling is involved in diencephalic development at late stages of embryonic development, but its roles and mechanisms of action during early neural development are still unclear. Here we demonstrate that mice lacking enzymatic activity of the acetyltransferase GCN5 ((Gcn5hat/hat )), which were previously characterized with respect to their exencephalic phenotype, exhibit significant diencephalic expansion, decreased diencephalic RA signaling, and increased diencephalic WNT and SHH signaling. Using a variety of molecular biology techniques in both cultured neuroepithelial cells treated with a GCN5 inhibitor and forebrain tissue from (Gcn5hat/hat ) embryos, we demonstrate that GCN5, RARα/γ, and the poorly characterized protein TACC1 form a complex in the nucleus that binds specific retinoic acid response elements in the absence of RA. Furthermore, RA triggers GCN5-mediated acetylation of TACC1, which results in dissociation of TACC1 from retinoic acid response elements and leads to transcriptional activation of RA target genes. Intriguingly, RA signaling defects caused by in vitro inhibition of GCN5 can be rescued through RA-dependent mechanisms that require RARβ. Last, we demonstrate that the diencephalic expansion and transcriptional defects seen in (Gcn5hat/hat ) mutants can be rescued with gestational RA supplementation, supporting a direct link between GCN5, TACC1, and RA signaling in the developing diencephalon. Together, our studies identify a novel, nonhistone substrate for GCN5 whose modification regulates a previously undescribed, tissue-specific mechanism of RA signaling that is required to restrict diencephalic size during early forebrain development.SIGNIFICANCE STATEMENT Changes in diencephalic size and shape, as well as SNPs associated with retinoic acid (RA) signaling-associated genes, have been linked to neuropsychiatric disorders. However, the mechanisms that regulate diencephalic morphogenesis and the involvement of RA signaling in this process are poorly understood. Here we demonstrate a novel role of the acetyltransferase GCN5 in a previously undescribed mechanism of RA signaling in the developing forebrain that is required to maintain the appropriate size of the diencephalon. Together, our experiments identify a novel nonhistone substrate of GCN5, highlight an essential role for both GCN5 and RA signaling in early diencephalic development, and elucidate a novel molecular regulatory mechanism for RA signaling that is specific to the developing forebrain.
Collapse
|
20
|
Sena E, Feistel K, Durand BC. An Evolutionarily Conserved Network Mediates Development of the zona limitans intrathalamica, a Sonic Hedgehog-Secreting Caudal Forebrain Signaling Center. J Dev Biol 2016; 4:jdb4040031. [PMID: 29615594 PMCID: PMC5831802 DOI: 10.3390/jdb4040031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/29/2016] [Accepted: 10/13/2016] [Indexed: 12/16/2022] Open
Abstract
Recent studies revealed new insights into the development of a unique caudal forebrain-signaling center: the zona limitans intrathalamica (zli). The zli is the last brain signaling center to form and the first forebrain compartment to be established. It is the only part of the dorsal neural tube expressing the morphogen Sonic Hedgehog (Shh) whose activity participates in the survival, growth and patterning of neuronal progenitor subpopulations within the thalamic complex. Here, we review the gene regulatory network of transcription factors and cis-regulatory elements that underlies formation of a shh-expressing delimitated domain in the anterior brain. We discuss evidence that this network predates the origin of chordates. We highlight the contribution of Shh, Wnt and Notch signaling to zli development and discuss implications for the fact that the morphogen Shh relies on primary cilia for signal transduction. The network that underlies zli development also contributes to thalamus induction, and to its patterning once the zli has been set up. We present an overview of the brain malformations possibly associated with developmental defects in this gene regulatory network (GRN).
Collapse
Affiliation(s)
- Elena Sena
- Institut Curie, Université Paris Sud, INSERM U1021, CNRS UMR3347, Centre Universitaire, Bâtiment 110, F-91405 Orsay Cedex, France.
| | - Kerstin Feistel
- Institute of Zoology, University of Hohenheim, Garbenstr. 30, 70593 Stuttgart, Germany.
| | - Béatrice C Durand
- Institut Curie, Université Paris Sud, INSERM U1021, CNRS UMR3347, Centre Universitaire, Bâtiment 110, F-91405 Orsay Cedex, France.
| |
Collapse
|
21
|
Zhang Y, Alvarez-Bolado G. Differential developmental strategies by Sonic hedgehog in thalamus and hypothalamus. J Chem Neuroanat 2016; 75:20-7. [DOI: 10.1016/j.jchemneu.2015.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 11/25/2015] [Indexed: 12/11/2022]
|
22
|
Holland LZ. The origin and evolution of chordate nervous systems. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2015.0048. [PMID: 26554041 DOI: 10.1098/rstb.2015.0048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the past 40 years, comparisons of developmental gene expression and mechanisms of development (evodevo) joined comparative morphology as tools for reconstructing long-extinct ancestral forms. Unfortunately, both approaches typically give congruent answers only with closely related organisms. Chordate nervous systems are good examples. Classical studies alone left open whether the vertebrate brain was a new structure or evolved from the anterior end of an ancestral nerve cord like that of modern amphioxus. Evodevo plus electron microscopy showed that the amphioxus brain has a diencephalic forebrain, small midbrain, hindbrain and spinal cord with parts of the genetic mechanisms for the midbrain/hindbrain boundary, zona limitans intrathalamica and neural crest. Evodevo also showed how extra genes resulting from whole-genome duplications in vertebrates facilitated evolution of new structures like neural crest. Understanding how the chordate central nervous system (CNS) evolved from that of the ancestral deuterostome has been truly challenging. The majority view is that this ancestor had a CNS with a brain that gave rise to the chordate CNS and, with loss of a discrete brain, to one of the two hemichordate nerve cords. The minority view is that this ancestor had no nerve cord; those in chordates and hemichordates evolved independently. New techniques such as phylostratigraphy may help resolve this conundrum.
Collapse
Affiliation(s)
- Linda Z Holland
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093-0202, USA
| |
Collapse
|
23
|
Wnt1 signal determines the patterning of the diencephalic dorso-ventral axis. Brain Struct Funct 2015; 221:3693-708. [PMID: 26452989 DOI: 10.1007/s00429-015-1126-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022]
Abstract
The diencephalon is a complex brain area that derives from the caudal region of the prosencephalon. This structure is divided into four longitudinal neuroepithelial zones: roof, alar, basal and floor plates, which constitute its dorso-ventral (DV) columnar domains. Morphogenetic differences between alar and basal plates in the prosencephalon and mesencephalon contribute to the characteristic expansion of alar plate derivatives in the brain and the formation of the cephalic flexure. Although differential histogenesis among DV regions seems to be relevant in understanding structural and functional complexity of the brain, most of our knowledge about DV regionalization comes from the spinal cord development. Therefore, it seems of interest to study the molecular mechanisms that govern DV patterning in the diencephalon, the brain region where strong differences in size and complexity between alar and basal derivatives are evident in all vertebrates. Different morphogenetic signals, which induce specific progenitors fate to the neighboring epithelium, are involved in the spinal cord DV patterning. To study if Wnt1, one of these signaling molecules, has a role for the establishment of the diencephalic longitudinal domains, we carried out gain- and loss-of-function experiments, using mice and chick embryos. Our results demonstrated functional differences in the molecular mechanisms downstream of Wnt1 function in the diencephalon, in relation to the spinal cord. We further demonstrated that Bmp4 signal induces Wnt1 expression in the diencephalon, unraveling a new molecular regulatory code downstream of primary dorsalizing signals to control ventral regionalization in the diencephalon.
Collapse
|
24
|
Martinez-Ferre A, Lloret-Quesada C, Prakash N, Wurst W, Rubenstein JLR, Martinez S. Fgf15 regulates thalamic development by controlling the expression of proneural genes. Brain Struct Funct 2015; 221:3095-109. [PMID: 26311466 DOI: 10.1007/s00429-015-1089-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 07/21/2015] [Indexed: 01/01/2023]
Abstract
The establishment of the brain structural complexity requires a precisely orchestrated interplay between extrinsic and intrinsic signals modulating cellular mechanisms to guide neuronal differentiation. However, little is known about the nature of these signals in the diencephalon, a complex brain region that processes and relays sensory and motor information to and from the cerebral cortex and subcortical structures. Morphogenetic signals from brain organizers regulate histogenetic processes such as cellular proliferation, migration, and differentiation. Sonic hedgehog (Shh) in the key signal of the ZLI, identified as the diencephalic organizer. Fgf15, the mouse gene orthologous of human, chick, and zebrafish Fgf19, is induced by Shh signal and expressed in the diencephalic alar plate progenitors during histogenetic developmental stages. This work investigates the role of Fgf15 signal in diencephalic development. In the absence of Fgf15, the complementary expression pattern of proneural genes: Ascl1 and Nng2, is disrupted and the GABAergic thalamic cells do not differentiate; in addition dorsal thalamic progenitors failed to exit from the mitotic cycle and to differentiate into neurons. Therefore, our findings indicate that Fgf15 is the Shh downstream signal to control thalamic regionalization, neurogenesis, and neuronal differentiation by regulating the expression and mutual segregation of neurogenic and proneural regulatory genes.
Collapse
Affiliation(s)
- Almudena Martinez-Ferre
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, 30120, El Palmar, Murcia, Spain
| | - Cosme Lloret-Quesada
- Institute of Neurosciences, Miguel Hernández University, Spanish National Research Council, San Juan Campus, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Nilima Prakash
- Institute of Developmental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Technical University Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Technical University Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - John L R Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA, USA
| | - Salvador Martinez
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, 30120, El Palmar, Murcia, Spain. .,Institute of Neurosciences, Miguel Hernández University, Spanish National Research Council, San Juan Campus, 03550, Sant Joan d'Alacant, Alicante, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Valencia, Spain.
| |
Collapse
|
25
|
Martinez-Lopez JE, Moreno-Bravo JA, Madrigal MP, Martinez S, Puelles E. Mesencephalic basolateral domain specification is dependent on Sonic Hedgehog. Front Neuroanat 2015; 9:12. [PMID: 25741244 PMCID: PMC4330881 DOI: 10.3389/fnana.2015.00012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/27/2015] [Indexed: 11/20/2022] Open
Abstract
In the study of central nervous system morphogenesis, the identification of new molecular markers allows us to identify domains along the antero-posterior and dorso-ventral (DV) axes. In the past years, the alar and basal plates of the midbrain have been divided into different domains. The precise location of the alar-basal boundary is still under discussion. We have identified Barhl1, Nhlh1 and Six3 as appropriate molecular markers to the adjacent domains of this transition. The description of their expression patterns and the contribution to the different mesencephalic populations corroborated their role in the specification of these domains. We studied the influence of Sonic Hedgehog on these markers and therefore on the specification of these territories. The lack of this morphogen produced severe alterations in the expression pattern of Barhl1 and Nhlh1 with consequent misspecification of the basolateral (BL) domain. Six3 expression was apparently unaffected, however its distribution changed leading to altered basal domains. In this study we confirmed the localization of the alar-basal boundary dorsal to the BL domain and demonstrated that the development of the BL domain highly depends on Shh.
Collapse
Affiliation(s)
- Jesus E Martinez-Lopez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Consejo Superior de Investigaciones Científicas (UMH-CSIC) Alicante, Spain
| | - Juan A Moreno-Bravo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Consejo Superior de Investigaciones Científicas (UMH-CSIC) Alicante, Spain
| | - M Pilar Madrigal
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Consejo Superior de Investigaciones Científicas (UMH-CSIC) Alicante, Spain
| | - Salvador Martinez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Consejo Superior de Investigaciones Científicas (UMH-CSIC) Alicante, Spain ; Instituto Murciano de Investigacion Biomedica IMIB-Arrixaca (CIBERSAM) Murcia, Spain
| | - Eduardo Puelles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Consejo Superior de Investigaciones Científicas (UMH-CSIC) Alicante, Spain
| |
Collapse
|
26
|
Juraver-Geslin HA, Durand BC. Early development of the neural plate: new roles for apoptosis and for one of its main effectors caspase-3. Genesis 2015; 53:203-24. [PMID: 25619400 DOI: 10.1002/dvg.22844] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022]
Abstract
Despite its tremendous complexity, the vertebrate nervous system emerges from a homogenous layer of neuroepithelial cells, the neural plate. Its formation relies on the time- and space-controlled progression of developmental programs. Apoptosis is a biological process that removes superfluous and potentially dangerous cells and is implemented through the activation of a molecular pathway conserved during evolution. Apoptosis and an unconventional function of one of its main effectors, caspase-3, contribute to the patterning and growth of the neuroepithelium. Little is known about the intrinsic and extrinsic cues controlling activities of the apoptotic machinery during development. The BarH-like (Barhl) proteins are homeodomain-containing transcription factors. The observations in Caenorhabditis elegans, Xenopus, and mice document that Barhl proteins act in cell survival and as cell type-specific regulators of a caspase-3 function that limits neural progenitor proliferation. In this review, we discuss the roles and regulatory modes of the apoptotic machinery in the development of the neural plate. We focus on the Barhl2, the Sonic Hedgehog, and the Wnt pathways and their activities in neural progenitor survival and proliferation.
Collapse
Affiliation(s)
- Hugo A Juraver-Geslin
- Department of Basic Science, Craniofacial Biology, College of Dentistry, New York University, New York, New York
| | | |
Collapse
|
27
|
Magnani D, Morlé L, Hasenpusch-Theil K, Paschaki M, Jacoby M, Schurmans S, Durand B, Theil T. The ciliogenic transcription factor Rfx3 is required for the formation of the thalamocortical tract by regulating the patterning of prethalamus and ventral telencephalon. Hum Mol Genet 2015; 24:2578-93. [PMID: 25631876 DOI: 10.1093/hmg/ddv021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/21/2015] [Indexed: 01/08/2023] Open
Abstract
Primary cilia are complex subcellular structures that play key roles during embryogenesis by controlling the cellular response to several signaling pathways. Defects in the function and/or structure of primary cilia underlie a large number of human syndromes collectively referred to as ciliopathies. Often, ciliopathies are associated with mental retardation (MR) and malformation of the corpus callosum. However, the possibility of defects in other forebrain axon tracts, which could contribute to the cognitive disorders of these patients, has not been explored. Here, we investigate the formation of the corticothalamic/thalamocortical tracts in mice mutant for Rfx3, which regulates the expression of many genes involved in ciliogenesis and cilia function. Using DiI axon tracing and immunohistochemistry experiments, we show that some Rfx3(-/-) corticothalamic axons abnormally migrate toward the pial surface of the ventral telencephalon (VT). Some thalamocortical axons (TCAs) also fail to leave the diencephalon or abnormally project toward the amygdala. Moreover, the Rfx3(-/-) VT displays heterotopias containing attractive guidance cues and expressing the guidance molecules Slit1 and Netrin1. Finally, the abnormal projection of TCAs toward the amygdala is also present in mice carrying a mutation in the Inpp5e gene, which is mutated in Joubert Syndrome and which controls cilia signaling and stability. The presence of identical thalamocortical malformations in two independent ciliary mutants indicates a novel role for primary cilia in the formation of the corticothalamic/thalamocortical tracts by establishing the correct cellular environment necessary for its development.
Collapse
Affiliation(s)
- Dario Magnani
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Laurette Morlé
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Université Claude Bernard Lyon 1, Villeurbanne, Lyon F69622, France
| | - Kerstin Hasenpusch-Theil
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Marie Paschaki
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Université Claude Bernard Lyon 1, Villeurbanne, Lyon F69622, France
| | - Monique Jacoby
- Institute of Immunology, Centre de Recherche Public de la Santé/Laboratoire National de Santé, Luxembourg, Luxembourg and
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-Signal Transduction, GIGA B34, Université de Liège, Liège B-4000, Belgium
| | - Bénédicte Durand
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Université Claude Bernard Lyon 1, Villeurbanne, Lyon F69622, France
| | - Thomas Theil
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK,
| |
Collapse
|
28
|
Juraver-Geslin HA, Gómez-Skarmeta JL, Durand BC. The conserved barH-like homeobox-2 gene barhl2 acts downstream of orthodentricle-2 and together with iroquois-3 in establishment of the caudal forebrain signaling center induced by Sonic Hedgehog. Dev Biol 2014; 396:107-20. [PMID: 25281935 DOI: 10.1016/j.ydbio.2014.09.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 08/13/2014] [Accepted: 09/23/2014] [Indexed: 01/01/2023]
Abstract
In this study, we investigated the gene regulatory network that governs formation of the Zona limitans intrathalamica (ZLI), a signaling center that secretes Sonic Hedgehog (Shh) to control the growth and regionalization of the caudal forebrain. Using loss- and gain-of-function, explants and grafting experiments in amphibians, we demonstrate that barhl2 acts downstream of otx2 and together with the iroquois (irx)-3 gene in establishment of the ZLI compartment initiated by Shh influence. We find that the presumptive (pre)-ZLI domain expresses barhl2, otx2 and irx3, whereas the thalamus territory caudally bordering the pre-ZLI expresses barhl2, otx2 and irx1/2 and early on irx3. We demonstrate that Barhl2 activity is required for determination of the ZLI and thalamus fates and that within the p2 alar plate the ratio of Irx3 to Irx1/2 contributes to ZLI specification and size determination. We show that when continuously exposed to Shh, neuroepithelial cells coexpressing barhl2, otx2 and irx3 acquire two characteristics of the ZLI compartment-the competence to express shh and the ability to segregate from anterior neural plate cells. In contrast, neuroepithelial cells expressing barhl2, otx2 and irx1/2, are not competent to express shh. Noteworthy in explants, under Shh influence, ZLI-like cells segregate from thalamic-like cells. Our study establishes that Barhl2 activity plays a key role in p2 alar plate patterning, specifically ZLI formation, and provides new insights on establishment of the signaling center of the caudal forebrain.
Collapse
Affiliation(s)
- Hugo A Juraver-Geslin
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 46 rue d'Ulm 75005, Paris F-75005, France; INSERM, U1024, Paris F-75005, France; CNRS, UMR 8197, Paris F-75005, France; S1.7 46 rue d'Ulm, 75005 Paris, France
| | - José Luis Gómez-Skarmeta
- Centro Andaluz de Biología del Desarrollo-CSIC/Universidad Pablo de Olavide, Carretera de Utrera, Km1, 41013 Sevilla, Spain
| | - Béatrice C Durand
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, S1.7 46 rue d'Ulm 75005, Paris F-75005, France; INSERM, U1024, Paris F-75005, France; CNRS, UMR 8197, Paris F-75005, France; S1.7 46 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
29
|
Caballero IM, Manuel MN, Molinek M, Quintana-Urzainqui I, Mi D, Shimogori T, Price DJ. Cell-autonomous repression of Shh by transcription factor Pax6 regulates diencephalic patterning by controlling the central diencephalic organizer. Cell Rep 2014; 8:1405-18. [PMID: 25176648 PMCID: PMC4241467 DOI: 10.1016/j.celrep.2014.07.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 06/24/2014] [Accepted: 07/27/2014] [Indexed: 11/17/2022] Open
Abstract
During development, region-specific patterns of regulatory gene expression are controlled by signaling centers that release morphogens providing positional information to surrounding cells. Regulation of signaling centers themselves is therefore critical. The size and the influence of a Shh-producing forebrain organizer, the zona limitans intrathalamica (ZLI), are limited by Pax6. By studying mouse chimeras, we find that Pax6 acts cell autonomously to block Shh expression in cells around the ZLI. Immunoprecipitation and luciferase assays indicate that Pax6 can bind the Shh promoter and repress its function. An analysis of chimeras suggests that many of the regional gene expression pattern defects that occur in Pax6−/− diencephalic cells result from a non-cell-autonomous position-dependent defect of local intercellular signaling. Blocking Shh signaling in Pax6−/− mutants reverses major diencephalic patterning defects. We conclude that Pax6’s cell-autonomous repression of Shh expression around the ZLI is critical for many aspects of normal diencephalic patterning. Pax6 limits the effects of a forebrain organizer, the zona limitans intrathalamica Pax6 blocks diencephalic Shh expression cell autonomously Absence of Pax6 causes non-cell-autonomous diencephalic patterning defects Blocking Shh signaling in Pax6−/− mutants reverses diencephalic patterning defects
Collapse
Affiliation(s)
| | - Martine N Manuel
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Michael Molinek
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | | | - Da Mi
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | | | - David J Price
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
30
|
Winkler T, Mahoney EJ, Sinner D, Wylie CC, Dahia CL. Wnt signaling activates Shh signaling in early postnatal intervertebral discs, and re-activates Shh signaling in old discs in the mouse. PLoS One 2014; 9:e98444. [PMID: 24892825 PMCID: PMC4043533 DOI: 10.1371/journal.pone.0098444] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 05/02/2014] [Indexed: 11/28/2022] Open
Abstract
Intervertebral discs (IVDs) are strong fibrocartilaginous joints that connect adjacent vertebrae of the spine. As discs age they become prone to failure, with neurological consequences that are often severe. Surgical repair of discs treats the result of the disease, which affects as many as one in seven people, rather than its cause. An ideal solution would be to repair degenerating discs using the mechanisms of their normal differentiation. However, these mechanisms are poorly understood. Using the mouse as a model, we previously showed that Shh signaling produced by nucleus pulposus cells activates the expression of differentiation markers, and cell proliferation, in the postnatal IVD. In the present study, we show that canonical Wnt signaling is required for the expression of Shh signaling targets in the IVD. We also show that Shh and canonical Wnt signaling pathways are down-regulated in adult IVDs. Furthermore, this down-regulation is reversible, since re-activation of the Wnt or Shh pathways in older discs can re-activate molecular markers of the IVD that are lost with age. These data suggest that biological treatments targeting Wnt and Shh signaling pathways may be feasible as a therapeutic for degenerative disc disease.
Collapse
Affiliation(s)
- Tamara Winkler
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Eric J. Mahoney
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Debora Sinner
- The Perinatal Institute Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Christopher C. Wylie
- Emeritus Professor, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Chitra Lekha Dahia
- Tissue Engineering Regeneration and Repair Program, Hospital for Special Surgery, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Cavodeassi F. Integration of anterior neural plate patterning and morphogenesis by the Wnt signaling pathway. Dev Neurobiol 2013; 74:759-71. [PMID: 24115566 DOI: 10.1002/dneu.22135] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 09/13/2013] [Accepted: 09/23/2013] [Indexed: 01/08/2023]
Abstract
Wnts are essential for a multitude of processes during embryonic development and adult homeostasis. The molecular structure of the Wnt pathway is extremely complex, and it keeps growing as new molecular components and novel interactions are uncovered. Recent studies have advanced our understanding on how the diverse molecular outcomes of the Wnt pathway are integrated during organ development, an integration that is also essential, although mechanistically poorly understood, during the formation of the anterior part of the nervous system, the forebrain. In this article, the author has summarized these findings and discussed their implications for forebrain development. A special emphasis has been put forth on studies performed in the zebrafish as this model system has been instrumental for our current understanding of forebrain patterning.
Collapse
Affiliation(s)
- Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, 28049, Madrid, Spain
| |
Collapse
|
32
|
Holland LZ, Carvalho JE, Escriva H, Laudet V, Schubert M, Shimeld SM, Yu JK. Evolution of bilaterian central nervous systems: a single origin? EvoDevo 2013; 4:27. [PMID: 24098981 PMCID: PMC3856589 DOI: 10.1186/2041-9139-4-27] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/14/2013] [Indexed: 12/21/2022] Open
Abstract
The question of whether the ancestral bilaterian had a central nervous system (CNS) or a diffuse ectodermal nervous system has been hotly debated. Considerable evidence supports the theory that a CNS evolved just once. However, an alternative view proposes that the chordate CNS evolved from the ectodermal nerve net of a hemichordate-like ancestral deuterostome, implying independent evolution of the CNS in chordates and protostomes. To specify morphological divisions along the anterior/posterior axis, this ancestor used gene networks homologous to those patterning three organizing centers in the vertebrate brain: the anterior neural ridge, the zona limitans intrathalamica and the isthmic organizer, and subsequent evolution of the vertebrate brain involved elaboration of these ancestral signaling centers; however, all or part of these signaling centers were lost from the CNS of invertebrate chordates. The present review analyzes the evidence for and against these theories. The bulk of the evidence indicates that a CNS evolved just once - in the ancestral bilaterian. Importantly, in both protostomes and deuterostomes, the CNS represents a portion of a generally neurogenic ectoderm that is internalized and receives and integrates inputs from sensory cells in the remainder of the ectoderm. The expression patterns of genes involved in medio/lateral (dorso/ventral) patterning of the CNS are similar in protostomes and chordates; however, these genes are not similarly expressed in the ectoderm outside the CNS. Thus, their expression is a better criterion for CNS homologs than the expression of anterior/posterior patterning genes, many of which (for example, Hox genes) are similarly expressed both in the CNS and in the remainder of the ectoderm in many bilaterians. The evidence leaves hemichordates in an ambiguous position - either CNS centralization was lost to some extent at the base of the hemichordates, or even earlier, at the base of the hemichordates + echinoderms, or one of the two hemichordate nerve cords is homologous to the CNS of protostomes and chordates. In any event, the presence of part of the genetic machinery for the anterior neural ridge, the zona limitans intrathalamica and the isthmic organizer in invertebrate chordates together with similar morphology indicates that these organizers were present, at least in part, at the base of the chordates and were probably elaborated upon in the vertebrate lineage.
Collapse
Affiliation(s)
- Linda Z Holland
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California at San Diego, La Jolla, CA 92093-0202, USA
| | - João E Carvalho
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009 – CNRS/UPMC), Observatoire Océanologique de Villefranche-sur-Mer, 181 Chemin du Lazaret, B.P. 28, 06230 Villefranche-sur-Mer, France
| | - Hector Escriva
- CNRS, UMR 7232, BIOM, Université Pierre et Marie Curie Paris 06, Observatoire Océanologique, 66650 Banyuls-sur-Mer, France
| | - Vincent Laudet
- Institut de Génomique Fonctionnelle de Lyon (CNRS UMR5242, UCBL, ENS, INRA 1288), Ecole Normale Supérieure de Lyon, 46 allée d’Italie, 69364 Lyon Cedex 07, France
| | - Michael Schubert
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009 – CNRS/UPMC), Observatoire Océanologique de Villefranche-sur-Mer, 181 Chemin du Lazaret, B.P. 28, 06230 Villefranche-sur-Mer, France
| | - Sebastian M Shimeld
- Department of Zoology, University of Oxford, The Tinbergen Building, South Parks Road, Oxford OX1 3PS, UK
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
33
|
Nicoleau C, Varela C, Bonnefond C, Maury Y, Bugi A, Aubry L, Viegas P, Bourgois-Rocha F, Peschanski M, Perrier AL. Embryonic stem cells neural differentiation qualifies the role of Wnt/β-Catenin signals in human telencephalic specification and regionalization. Stem Cells 2013; 31:1763-74. [DOI: 10.1002/stem.1462] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 04/21/2013] [Accepted: 05/11/2013] [Indexed: 02/05/2023]
Affiliation(s)
- Camille Nicoleau
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| | | | | | - Yves Maury
- CECS; I-STEM, AFM, Evry 91030 Cedex France
| | | | - Laetitia Aubry
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| | - Pedro Viegas
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| | - Fany Bourgois-Rocha
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| | - Marc Peschanski
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| | - Anselme L Perrier
- Inserm U861; I-STEM, AFM, Evry 91030 Cedex France
- UEVE U861; I-STEM, AFM, Evry 91030 Cedex France
| |
Collapse
|
34
|
Martinez-Ferre A, Navarro-Garberí M, Bueno C, Martinez S. Implantation of Dkk-1-soaked Beads into the Neural Tube of Chicken Embryos. Bio Protoc 2013. [DOI: 10.21769/bioprotoc.963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|