1
|
Qin H, Liang T, Zhang C, Wu J, Sheng X. The bidirectional relationship between cilia and PCP signaling pathway core protein Vangl2. Sci Prog 2025; 108:368504241311964. [PMID: 39819247 PMCID: PMC11748379 DOI: 10.1177/00368504241311964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Vangl2, a core component of the PCP signaling pathway, serves as a scaffold protein on the cell membrane, playing a crucial role in organizing protein complexes. Cilia, dynamic structures on the cell surface, carry out a wide range of functions. Research has highlighted a bidirectional regulatory interaction between Vangl2 and cilia, underscoring their interconnected roles in cellular processes. This relationship is demonstrated by the localization of Vangl2 at the base and proximal regions of cilia, where it plays essential roles in ciliary positioning, asymmetric distribution, and ciliogenesis. In contrast, the absence of cilia can disrupt Vangl2-mediated signal transduction processes. This review offers a narrative review of recent research on Vangl2's function in cilia and examines the regulatory effects of cilia on Vangl2-mediated signaling.
Collapse
Affiliation(s)
- Huanyong Qin
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting Liang
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Chuanfen Zhang
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Junlin Wu
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin Sheng
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
2
|
Wang S, Kang Y, Xie H. PKD2: An Important Membrane Protein in Organ Development. Cells 2024; 13:1722. [PMID: 39451240 PMCID: PMC11506562 DOI: 10.3390/cells13201722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
PKD2 was first identified as the pathogenic protein for autosomal dominant polycystic kidney disease (ADPKD) and is widely recognized as an ion channel. Subsequent studies have shown that PKD2 is widely expressed in various animal tissues and plays a crucial role in tissue and organ development. Additionally, PKD2 is conserved from single-celled organisms to vertebrates. Here, we provide an overview of recent advances in the function of PKD2 in key model animals, focusing on the establishment of left-right organ asymmetry, renal homeostasis, cardiovascular development, and signal transduction in reproduction and mating. We specifically focus on the roles of PKD2 in development and highlight future prospects for PKD2 research.
Collapse
Affiliation(s)
- Shuo Wang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Haibo Xie
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
3
|
Groh AMR, Song YL, Tea F, Lu B, Huynh S, Afanasiev E, Bigotte M, Del Bigio MR, Stratton JA. Multiciliated ependymal cells: an update on biology and pathology in the adult brain. Acta Neuropathol 2024; 148:39. [PMID: 39254862 DOI: 10.1007/s00401-024-02784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Mature multiciliated ependymal cells line the cerebral ventricles where they form a partial barrier between the cerebrospinal fluid (CSF) and brain parenchyma and regulate local CSF microcirculation through coordinated ciliary beating. Although the ependyma is a highly specialized brain interface with barrier, trophic, and perhaps even regenerative capacity, it remains a misfit in the canon of glial neurobiology. We provide an update to seminal reviews in the field by conducting a scoping review of the post-2010 mature multiciliated ependymal cell literature. We delineate how recent findings have either called into question or substantiated classical views of the ependymal cell. Beyond this synthesis, we document the basic methodologies and study characteristics used to describe multiciliated ependymal cells since 1980. Our review serves as a comprehensive resource for future investigations of mature multiciliated ependymal cells.
Collapse
Affiliation(s)
- Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Yeji Lori Song
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Fiona Tea
- Department of Neuroscience, University of Montreal, Montréal, QC, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Stephanie Huynh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Marc R Del Bigio
- Department of Pathology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
4
|
Ross KG, Alvarez Zepeda S, Auwal MA, Garces AK, Roman S, Zayas RM. The Role of Polycystic Kidney Disease-Like Homologs in Planarian Nervous System Regeneration and Function. Integr Org Biol 2024; 6:obae035. [PMID: 39364443 PMCID: PMC11448475 DOI: 10.1093/iob/obae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
Planarians are an excellent model for investigating molecular mechanisms necessary for regenerating a functional nervous system. Numerous studies have led to the generation of extensive genomic resources, especially whole-animal single-cell RNA-seq resources. These have facilitated in silico predictions of neuronal subtypes, many of which have been anatomically mapped by in situ hybridization. However, our knowledge of the function of dozens of neuronal subtypes remains poorly understood. Previous investigations identified that polycystic kidney disease (pkd)-like genes in planarians are strongly expressed in sensory neurons and have roles in mechanosensation. Here, we examine the expression and function of all the pkd genes found in the Schmidtea mediterranea genome and map their expression in the asexual and hermaphroditic strains. Using custom behavioral assays, we test the function of pkd genes in response to mechanical stimulation and in food detection. Our work provides insight into the physiological function of sensory neuron populations and protocols for creating inexpensive automated setups for acquiring and analyzing mechanosensory stimulation in planarians.
Collapse
Affiliation(s)
- K G Ross
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - S Alvarez Zepeda
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - M A Auwal
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - A K Garces
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - S Roman
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - R M Zayas
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| |
Collapse
|
5
|
Donati A, Schneider-Maunoury S, Vesque C. Centriole Translational Planar Polarity in Monociliated Epithelia. Cells 2024; 13:1403. [PMID: 39272975 PMCID: PMC11393834 DOI: 10.3390/cells13171403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Ciliated epithelia are widespread in animals and play crucial roles in many developmental and physiological processes. Epithelia composed of multi-ciliated cells allow for directional fluid flow in the trachea, oviduct and brain cavities. Monociliated epithelia play crucial roles in vertebrate embryos, from the establishment of left-right asymmetry to the control of axis curvature via cerebrospinal flow motility in zebrafish. Cilia also have a central role in the motility and feeding of free-swimming larvae in a variety of marine organisms. These diverse functions rely on the coordinated orientation (rotational polarity) and asymmetric localization (translational polarity) of cilia and of their centriole-derived basal bodies across the epithelium, both being forms of planar cell polarity (PCP). Here, we review our current knowledge on the mechanisms of the translational polarity of basal bodies in vertebrate monociliated epithelia from the molecule to the whole organism. We highlight the importance of live imaging for understanding the dynamics of centriole polarization. We review the roles of core PCP pathways and of apicobasal polarity proteins, such as Par3, whose central function in this process has been recently uncovered. Finally, we emphasize the importance of the coordination between polarity proteins, the cytoskeleton and the basal body itself in this highly dynamic process.
Collapse
Affiliation(s)
- Antoine Donati
- Developmental Biology Unit, UMR7622, Institut de Biologie Paris Seine (IBPS), Sorbonne Université, CNRS, INSERM U1156, 75005 Paris, France
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sylvie Schneider-Maunoury
- Developmental Biology Unit, UMR7622, Institut de Biologie Paris Seine (IBPS), Sorbonne Université, CNRS, INSERM U1156, 75005 Paris, France
| | - Christine Vesque
- Developmental Biology Unit, UMR7622, Institut de Biologie Paris Seine (IBPS), Sorbonne Université, CNRS, INSERM U1156, 75005 Paris, France
| |
Collapse
|
6
|
Kamalian A, Shirzadeh Barough S, Ho SG, Albert M, Luciano MG, Yasar S, Moghekar A. Molecular signatures of normal pressure hydrocephalus: a large-scale proteomic analysis of cerebrospinal fluid. Fluids Barriers CNS 2024; 21:64. [PMID: 39118132 PMCID: PMC11312837 DOI: 10.1186/s12987-024-00561-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Given the persistent challenge of differentiating idiopathic Normal Pressure Hydrocephalus (iNPH) from similar clinical entities, we conducted an in-depth proteomic study of cerebrospinal fluid (CSF) in 28 shunt-responsive iNPH patients, 38 Mild Cognitive Impairment (MCI) due to Alzheimer's disease, and 49 healthy controls. Utilizing the Olink Explore 3072 panel, we identified distinct proteomic profiles in iNPH that highlight significant downregulation of synaptic markers and cell-cell adhesion proteins. Alongside vimentin and inflammatory markers upregulation, these results suggest ependymal layer and transependymal flow dysfunction. Moreover, downregulation of multiple proteins associated with congenital hydrocephalus (e.g., L1CAM, PCDH9, ISLR2, ADAMTSL2, and B4GAT1) points to a possible shared molecular foundation between congenital hydrocephalus and iNPH. Through orthogonal partial least squares discriminant analysis (OPLS-DA), a panel comprising 13 proteins has been identified as potential diagnostic biomarkers of iNPH, pending external validation. These findings offer novel insights into the pathophysiology of iNPH, with implications for improved diagnosis.
Collapse
Affiliation(s)
- Aida Kamalian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | | | - Sara G Ho
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Mark G Luciano
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Sevil Yasar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA.
| |
Collapse
|
7
|
Ross KG, Zepeda SA, Auwal MA, Garces AK, Roman S, Zayas RM. The role of polycystic kidney disease-like homologs in planarian nervous system regeneration and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603829. [PMID: 39091889 PMCID: PMC11291080 DOI: 10.1101/2024.07.17.603829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Planarians are an excellent model for investigating molecular mechanisms necessary for regenerating a functional nervous system. Numerous studies have led to the generation of extensive genomic resources, especially whole-animal single-cell RNA-seq resources. These have facilitated in silico predictions of neuronal subtypes, many of which have been anatomically mapped by in situ hybridization. However, our knowledge of the function of dozens of neuronal subtypes remains poorly understood. Previous investigations identified that polycystic kidney disease (pkd)-like genes in planarians are strongly expressed in sensory neurons and have roles in mechanosensation. Here, we examine the expression and function of all the pkd genes found in the Schmidtea mediterranea genome and map their expression in the asexual and hermaphroditic strains. Using custom behavioral assays, we test the function of pkd genes in response to mechanical stimulation and in food detection. Our work provides insight into the physiological function of sensory neuron populations and protocols for creating inexpensive automated setups for acquiring and analyzing mechanosensory stimulation in planarians.
Collapse
Affiliation(s)
- Kelly G. Ross
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Sarai Alvarez Zepeda
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Mohammad A. Auwal
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Audrey K. Garces
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Sydney Roman
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Ricardo M. Zayas
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| |
Collapse
|
8
|
Kamalian A, Barough SS, Ho SG, Albert M, Luciano MG, Yasar S, Moghekar A. Molecular Signatures of Normal Pressure Hydrocephalus: A Largescale Proteomic Analysis of Cerebrospinal Fluid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.583014. [PMID: 38496536 PMCID: PMC10942380 DOI: 10.1101/2024.03.01.583014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Given the persistent challenge of differentiating idiopathic Normal Pressure Hydrocephalus (iNPH) from similar clinical entities, we conducted an in-depth proteomic study of cerebrospinal fluid (CSF) in 28 shunt-responsive iNPH patients, 38 Mild Cognitive Impairment (MCI) due to Alzheimer's disease, and 49 healthy controls. Utilizing the Olink Explore 3072 panel, we identified distinct proteomic profiles in iNPH that highlight significant downregulation of synaptic markers and cell-cell adhesion proteins. Alongside vimentin and inflammatory markers upregulation, these results suggest ependymal layer and transependymal flow dysfunction. Moreover, downregulation of multiple proteins associated with congenital hydrocephalus (e.g., L1CAM, PCDH9, ISLR2, ADAMTSL2, and B4GAT1) points to a possible shared molecular foundation between congenital hydrocephalus and iNPH. Through orthogonal partial least squares discriminant analysis (OPLS-DA), a panel comprising 13 proteins has been identified as potential diagnostic biomarkers of iNPH, pending external validation. These findings offer novel insights into the pathophysiology of iNPH, with implications for improved diagnosis.
Collapse
Affiliation(s)
- Aida Kamalian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | - Sara G. Ho
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Mark G. Luciano
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Sevil Yasar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| |
Collapse
|
9
|
Nualart F, Cifuentes M, Ramírez E, Martínez F, Barahona MJ, Ferrada L, Saldivia N, Bongarzone ER, Thorens B, Salazar K. Hyperglycemia increases SCO-spondin and Wnt5a secretion into the cerebrospinal fluid to regulate ependymal cell beating and glucose sensing. PLoS Biol 2023; 21:e3002308. [PMID: 37733692 PMCID: PMC10513282 DOI: 10.1371/journal.pbio.3002308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Hyperglycemia increases glucose concentrations in the cerebrospinal fluid (CSF), activating glucose-sensing mechanisms and feeding behavior in the hypothalamus. Here, we discuss how hyperglycemia temporarily modifies ependymal cell ciliary beating to increase hypothalamic glucose sensing. A high level of glucose in the rat CSF stimulates glucose transporter 2 (GLUT2)-positive subcommissural organ (SCO) cells to release SCO-spondin into the dorsal third ventricle. Genetic inactivation of mice GLUT2 decreases hyperglycemia-induced SCO-spondin secretion. In addition, SCO cells secrete Wnt5a-positive vesicles; thus, Wnt5a and SCO-spondin are found at the apex of dorsal ependymal cilia to regulate ciliary beating. Frizzled-2 and ROR2 receptors, as well as specific proteoglycans, such as glypican/testican (essential for the interaction of Wnt5a with its receptors) and Cx43 coupling, were also analyzed in ependymal cells. Finally, we propose that the SCO-spondin/Wnt5a/Frizzled-2/Cx43 axis in ependymal cells regulates ciliary beating, a cyclic and adaptive signaling mechanism to control glucose sensing.
Collapse
Affiliation(s)
- Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Manuel Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga Biomedical Research Institute and Nanomedicine Platform (IBIMA-BIONAND Platform), Malaga, Spain
| | - Eder Ramírez
- Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Fernando Martínez
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - María José Barahona
- Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Luciano Ferrada
- Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Natalia Saldivia
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Katterine Salazar
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| |
Collapse
|
10
|
Sunadome K, Erickson AG, Kah D, Fabry B, Adori C, Kameneva P, Faure L, Kanatani S, Kaucka M, Dehnisch Ellström I, Tesarova M, Zikmund T, Kaiser J, Edwards S, Maki K, Adachi T, Yamamoto T, Fried K, Adameyko I. Directionality of developing skeletal muscles is set by mechanical forces. Nat Commun 2023; 14:3060. [PMID: 37244931 PMCID: PMC10224984 DOI: 10.1038/s41467-023-38647-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/05/2023] [Indexed: 05/29/2023] Open
Abstract
Formation of oriented myofibrils is a key event in musculoskeletal development. However, the mechanisms that drive myocyte orientation and fusion to control muscle directionality in adults remain enigmatic. Here, we demonstrate that the developing skeleton instructs the directional outgrowth of skeletal muscle and other soft tissues during limb and facial morphogenesis in zebrafish and mouse. Time-lapse live imaging reveals that during early craniofacial development, myoblasts condense into round clusters corresponding to future muscle groups. These clusters undergo oriented stretch and alignment during embryonic growth. Genetic perturbation of cartilage patterning or size disrupts the directionality and number of myofibrils in vivo. Laser ablation of musculoskeletal attachment points reveals tension imposed by cartilage expansion on the forming myofibers. Application of continuous tension using artificial attachment points, or stretchable membrane substrates, is sufficient to drive polarization of myocyte populations in vitro. Overall, this work outlines a biomechanical guidance mechanism that is potentially useful for engineering functional skeletal muscle.
Collapse
Affiliation(s)
- Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Alek G Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Delf Kah
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Csaba Adori
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
- Department of Molecular Biosciences, Wenner Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Polina Kameneva
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Marketa Kaucka
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Str.2, 24306, Plön, Germany
| | | | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Steven Edwards
- KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Koichiro Maki
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, 606-8501, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden.
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria.
| |
Collapse
|
11
|
LRP2 contributes to planar cell polarity-dependent coordination of motile cilia function. Cell Tissue Res 2023; 392:535-551. [PMID: 36764939 PMCID: PMC10172251 DOI: 10.1007/s00441-023-03757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 02/12/2023]
Abstract
Motile cilia are protruding organelles on specialized epithelia that beat in a synchronous fashion to propel extracellular fluids. Coordination and orientation of cilia beating on individual cells and across tissues is a complex process dependent on planar cell polarity (PCP) signaling. Asymmetric sorting of PCP pathway components, essential to establish planar polarity, involves trafficking along the endocytic path, but the underlying regulatory processes remain incompletely understood. Here, we identified the endocytic receptor LRP2 as regulator of PCP component trafficking in ependyma, a multi-ciliated cell type that is involved in facilitating flow of the cerebrospinal fluid in the brain ventricular system. Lack of receptor expression in gene-targeted mice results in a failure to sort PCP core proteins to the anterior or posterior cell side and, consequently, in the inability to coordinate cilia arrangement and to aligned beating (loss of rotational and translational polarity). LRP2 deficiency coincides with a failure to sort NHERF1, a cytoplasmic LRP2 adaptor to the anterior cell side. As NHERF1 is essential to translocate PCP core protein Vangl2 to the plasma membrane, these data suggest a molecular mechanism whereby LRP2 interacts with PCP components through NHERF1 to control their asymmetric sorting along the endocytic path. Taken together, our findings identified the endocytic receptor LRP2 as a novel regulator of endosomal trafficking of PCP proteins, ensuring their asymmetric partition and establishment of translational and rotational planar cell polarity in the ependyma.
Collapse
|
12
|
Karimy JK, Newville JC, Sadegh C, Morris JA, Monuki ES, Limbrick DD, McAllister Ii JP, Koschnitzky JE, Lehtinen MK, Jantzie LL. Outcomes of the 2019 hydrocephalus association workshop, "Driving common pathways: extending insights from posthemorrhagic hydrocephalus". Fluids Barriers CNS 2023; 20:4. [PMID: 36639792 PMCID: PMC9838022 DOI: 10.1186/s12987-023-00406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The Hydrocephalus Association (HA) workshop, Driving Common Pathways: Extending Insights from Posthemorrhagic Hydrocephalus, was held on November 4 and 5, 2019 at Washington University in St. Louis. The workshop brought together a diverse group of basic, translational, and clinical scientists conducting research on multiple hydrocephalus etiologies with select outside researchers. The main goals of the workshop were to explore areas of potential overlap between hydrocephalus etiologies and identify drug targets that could positively impact various forms of hydrocephalus. This report details the major themes of the workshop and the research presented on three cell types that are targets for new hydrocephalus interventions: choroid plexus epithelial cells, ventricular ependymal cells, and immune cells (macrophages and microglia).
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Family Medicine, Mountain Area Health Education Center - Boone, North Carolina, 28607, USA
| | - Jessie C Newville
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Cameron Sadegh
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, MA, Boston, 02114, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jill A Morris
- National Institute of Neurological Disorders and Stroke, Neuroscience Center, National Institutes of Health, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD, 20892, USA
| | - Edwin S Monuki
- Departments of Pathology & Laboratory Medicine and Developmental & Cell Biology, University of California Irvine, Irvine, CA, 92697, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - James P McAllister Ii
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | | | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Lauren L Jantzie
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
- Kennedy Krieger Institute, Baltimore, MD, 21287, USA.
| |
Collapse
|
13
|
Olovnikov AM. Planetary Metronome as a Regulator of Lifespan and Aging Rate: The Metronomic Hypothesis. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1640-1650. [PMID: 36717453 DOI: 10.1134/s0006297922120197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A metronomic mechanism for the duration control of ontogenetic cycle periods of an animal is proposed. The components of the proposed metronomic system include the ventricular system of the brain, planet Earth as a generator of metronomic signals, and temporal DNA (tDNA) as a substrate that is epigenetically marked to measure elapsed time of ontogenesis. The metronomic system generates repetitive signals in the form of hydrodynamic disturbances in the cerebrospinal fluid (CSF). The metronomic effect arises due to the superposition of two processes - the near-wall unidirectional flow of CSF and oscillations in the movement of the planet. Hydrodynamic impacts of the metronome are transformed into nerve impulses that initiate epigenetic modification of tDNA in neurons, changing the content of factors expressed by this DNA for innervated targets of the body. The duration of ontogenetic cycle periods, including duration of the adult life, depends on the rate of addition of epigenetic marks to tDNA. This rate depends mainly on the frequency of the metronomic signals used by each particular species. But epigenetic modifications can also be influenced by factors that modulate metabolism and the rate of chromatin modifications, such as a calorie-restricted diet.
Collapse
Affiliation(s)
- Alexey M Olovnikov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| |
Collapse
|
14
|
Herranz-Pérez V, Nakatani J, Ishii M, Katada T, García-Verdugo JM, Ohata S. Ependymoma associated protein Zfta is expressed in immature ependymal cells but is not essential for ependymal development in mice. Sci Rep 2022; 12:1493. [PMID: 35087169 PMCID: PMC8795269 DOI: 10.1038/s41598-022-05526-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
The fusion protein of uncharacterised zinc finger translocation associated (ZFTA) and effector transcription factor of tumorigenic NF-κB signalling, RELA (ZFTA-RELA), is expressed in more than two-thirds of supratentorial ependymoma (ST-EPN-RELA), but ZFTA's expression profile and functional analysis in multiciliated ependymal (E1) cells have not been examined. Here, we showed the mRNA expression of mouse Zfta peaks on embryonic day (E) 17.5 in the wholemount of the lateral walls of the lateral ventricle. Zfta was expressed in the nuclei of FoxJ1-positive immature E1 (pre-E1) cells in E18.5 mouse embryonic brain. Interestingly, the transcription factors promoting ciliogenesis (ciliary TFs) (e.g., multicilin) and ZFTA-RELA upregulated luciferase activity using a 5' upstream sequence of ZFTA in cultured cells. Zftatm1/tm1 knock-in mice did not show developmental defects or abnormal fertility. In the Zftatm1/tm1 E1 cells, morphology, gene expression, ciliary beating frequency and ependymal flow were unaffected. These results suggest that Zfta is expressed in pre-E1 cells, possibly under the control of ciliary TFs, but is not essential for ependymal development or flow. This study sheds light on the mechanism of the ZFTA-RELA expression in the pathogenesis of ST-EPN-RELA: Ciliary TFs initiate ZFTA-RELA expression in pre-E1 cells, and ZFTA-RELA enhances its own expression using positive feedback.
Collapse
Affiliation(s)
- Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, CIBERNED, University of Valencia, 46980, Paterna, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, 46100, Burjassot, Spain
| | - Jin Nakatani
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Masaki Ishii
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Toshiaki Katada
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Jose Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, CIBERNED, University of Valencia, 46980, Paterna, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, 46100, Burjassot, Spain
| | - Shinya Ohata
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan.
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
15
|
Ludwig HC, Dreha-Kulaczewski S, Bock HC. Neurofluids-Deep inspiration, cilia and preloading of the astrocytic network. J Neurosci Res 2021; 99:2804-2821. [PMID: 34323313 DOI: 10.1002/jnr.24935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/15/2021] [Accepted: 07/13/2021] [Indexed: 01/20/2023]
Abstract
With the advent of real-time MRI, the motion and passage of cerebrospinal fluid can be visualized without gating and exclusion of low-frequency waves. This imaging modality gives insights into low-volume, rapidly oscillating cardiac-driven movement as well as sustained, high-volume, slowly oscillating inspiration-driven movement. Inspiration means a spontaneous or artificial increase in the intrathoracic dimensions independent of body position. Alterations in thoracic diameter enable the thoracic and spinal epidural venous compartments to be emptied and filled, producing an upward surge of cerebrospinal fluid inside the spine during inspiration; this surge counterbalances the downward pooling of venous blood toward the heart. Real-time MRI, as a macroscale in vivo observation method, could expand our knowledge of neurofluid dynamics, including how astrocytic fluid preloading is adjusted and how brain buoyancy and turgor are maintained in different postures and zero gravity. Along with these macroscale findings, new microscale insights into aquaporin-mediated fluid transfer, its sensing by cilia, and its tuning by nitric oxide will be reviewed. By incorporating clinical knowledge spanning several disciplines, certain disorders-congenital hydrocephalus with Chiari malformation, idiopathic intracranial hypertension, and adult idiopathic hydrocephalus-are interpreted and reviewed according to current concepts, from the basics of the interrelated systems to their pathology.
Collapse
Affiliation(s)
- Hans C Ludwig
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Steffi Dreha-Kulaczewski
- Division of Pediatric Neurology, Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Hans C Bock
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
16
|
Cumulative Damage: Cell Death in Posthemorrhagic Hydrocephalus of Prematurity. Cells 2021; 10:cells10081911. [PMID: 34440681 PMCID: PMC8393895 DOI: 10.3390/cells10081911] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 12/19/2022] Open
Abstract
Globally, approximately 11% of all infants are born preterm, prior to 37 weeks’ gestation. In these high-risk neonates, encephalopathy of prematurity (EoP) is a major cause of both morbidity and mortality, especially for neonates who are born very preterm (<32 weeks gestation). EoP encompasses numerous types of preterm birth-related brain abnormalities and injuries, and can culminate in a diverse array of neurodevelopmental impairments. Of note, posthemorrhagic hydrocephalus of prematurity (PHHP) can be conceptualized as a severe manifestation of EoP. PHHP impacts the immature neonatal brain at a crucial timepoint during neurodevelopment, and can result in permanent, detrimental consequences to not only cerebrospinal fluid (CSF) dynamics, but also to white and gray matter development. In this review, the relevant literature related to the diverse mechanisms of cell death in the setting of PHHP will be thoroughly discussed. Loss of the epithelial cells of the choroid plexus, ependymal cells and their motile cilia, and cellular structures within the glymphatic system are of particular interest. Greater insights into the injuries, initiating targets, and downstream signaling pathways involved in excess cell death shed light on promising areas for therapeutic intervention. This will bolster current efforts to prevent, mitigate, and reverse the consequential brain remodeling that occurs as a result of hydrocephalus and other components of EoP.
Collapse
|
17
|
Donati A, Anselme I, Schneider-Maunoury S, Vesque C. Planar polarization of cilia in the zebrafish floor-plate involves Par3-mediated posterior localization of highly motile basal bodies. Development 2021; 148:269080. [PMID: 34104942 DOI: 10.1242/dev.196386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Epithelial cilia, whether motile or primary, often display an off-center planar localization within the apical cell surface. This form of planar cell polarity (PCP) involves the asymmetric positioning of the ciliary basal body (BB). Using the monociliated epithelium of the embryonic zebrafish floor-plate, we investigated the dynamics and mechanisms of BB polarization by live imaging. BBs were highly motile, making back-and-forth movements along the antero-posterior (AP) axis and contacting both the anterior and posterior membranes. Contacts exclusively occurred at junctional Par3 patches and were often preceded by membrane digitations extending towards the BB, suggesting focused cortical pulling forces. Accordingly, BBs and Par3 patches were linked by dynamic microtubules. Later, BBs became less motile and eventually settled at posterior apical junctions enriched in Par3. BB posterior positioning followed Par3 posterior enrichment and was impaired upon Par3 depletion or disorganization of Par3 patches. In the PCP mutant vangl2, BBs were still motile but displayed poorly oriented membrane contacts that correlated with Par3 patch fragmentation and lateral spreading. Thus, we propose an unexpected function for posterior Par3 enrichment in controlling BB positioning downstream of the PCP pathway.
Collapse
Affiliation(s)
- Antoine Donati
- Sorbonne Université, CNRS UMR7622, INSERM U1156, Institut de Biologie Paris Seine (IBPS), Developmental Biology Unit, 75005 Paris, France
| | - Isabelle Anselme
- Sorbonne Université, CNRS UMR7622, INSERM U1156, Institut de Biologie Paris Seine (IBPS), Developmental Biology Unit, 75005 Paris, France
| | - Sylvie Schneider-Maunoury
- Sorbonne Université, CNRS UMR7622, INSERM U1156, Institut de Biologie Paris Seine (IBPS), Developmental Biology Unit, 75005 Paris, France
| | - Christine Vesque
- Sorbonne Université, CNRS UMR7622, INSERM U1156, Institut de Biologie Paris Seine (IBPS), Developmental Biology Unit, 75005 Paris, France
| |
Collapse
|
18
|
Yamada S, Ishikawa M, Nozaki K. Exploring mechanisms of ventricular enlargement in idiopathic normal pressure hydrocephalus: a role of cerebrospinal fluid dynamics and motile cilia. Fluids Barriers CNS 2021; 18:20. [PMID: 33874972 PMCID: PMC8056523 DOI: 10.1186/s12987-021-00243-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/13/2021] [Indexed: 11/15/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is considered an age-dependent chronic communicating hydrocephalus associated with cerebrospinal fluid (CSF) malabsorption; however, the aetiology of ventricular enlargement in iNPH has not yet been elucidated. There is accumulating evidence that support the hypothesis that various alterations in CSF dynamics contribute to ventricle dilatation in iNPH. This review focuses on CSF dynamics associated with ventriculomegaly and summarises the current literature based on three potential aetiology factors: genetic, environmental and hydrodynamic. The majority of gene mutations that cause communicating hydrocephalus were associated with an abnormal structure or dysfunction of motile cilia on the ventricular ependymal cells. Aging, alcohol consumption, sleep apnoea, diabetes and hypertension are candidates for the risk of developing iNPH, although there is no prospective cohort study to investigate the risk factors for iNPH. Alcohol intake may be associated with the dysfunction of ependymal cilia and sustained high CSF sugar concentration due to uncontrolled diabetes increases the fluid viscosity which in turn increases the shear stress on the ventricular wall surface. Sleep apnoea, diabetes and hypertension are known to be associated with the impairment of CSF and interstitial fluid exchange. Oscillatory shear stress to the ventricle wall surfaces is considerably increased by reciprocating bidirectional CSF movements in iNPH. Increased oscillatory shear stress impedes normal cilia beating, leading to motile cilia shedding from the ependymal cells. At the lack of ciliary protection, the ventricular wall is directly exposed to increased oscillatory shear stress. Additionally, increased oscillatory shear stress may be involved in activating the flow-mediated dilation signalling of the ventricular wall. In conclusion, as the CSF stroke volume at the cerebral aqueduct increases, the oscillatory shear stress increases, promoting motor cilia shedding and loss of ependymal cell coverage. These are considered to be the leading causes of ventricular enlargement in iNPH.
Collapse
Affiliation(s)
- Shigeki Yamada
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan. .,Interfaculty Initiative in Information Studies, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Masatsune Ishikawa
- Department of Neurosurgery and Normal Pressure Hydrocephalus Center, Rakuwakai Otowa Hospital, Kyoto, Japan.,Rakuwa Villa Ilios, Kyoto, Japan
| | - Kazuhiko Nozaki
- Department of Neurosurgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
19
|
Yoshida A, Kawata D, Shinotsuka N, Yoshida M, Yamaguchi Y, Miura M. Evidence for the involvement of caspases in establishing proper cerebrospinal fluid hydrodynamics. Neurosci Res 2021; 170:145-153. [PMID: 33417971 DOI: 10.1016/j.neures.2020.12.006] [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: 10/13/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 11/19/2022]
Abstract
A large number of cells undergo apoptosis via caspase activation during and after neural tube closure (NTC) in mammals. Apoptosis is executed by either intrinsic or extrinsic apoptotic pathways, and inhibition of each pathway causes developmental defects around NTC stages, which hampers the physiological roles of apoptosis and caspases after NTC. We generated transgenic mice in which a broad spectrum of caspases could be suppressed in a spatiotemporal manner by pan-caspase inhibitor protein p35 originating from baculovirus. Mice with nervous system-specific expression of p35 (Nestin-Cre (NCre);p35V mice) exhibited postnatal lethality within 1 month after birth. They were born at the expected Mendelian ratio, but demonstrated severe postnatal growth retardation and hydrocephalus. The flow of cerebrospinal fluid (CSF) between the third and fourth ventricles was disturbed, whereas neither stenosis nor abnormality in ciliary morphology was observed in the pathway of CSF flow. Hydrocephalus and growth retardation of NCre;p35V mice were not rescued by the deletion of RIPK3, an essential factor for necroptosis which occurs in the absence of caspase-8 activation during development. The CSF of NCre;p35V mice contained a larger amount of secreted proteins than that of the controls. These findings suggest that the establishment of proper CSF dynamics requires caspase activity during brain development after NTC.
Collapse
Affiliation(s)
- Ayako Yoshida
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Daisuke Kawata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naomi Shinotsuka
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mariko Yoshida
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Hibernation Metabolism, Physiology, and Development Group, Institute of Low Temperature Science, Hokkaido University, Sapporo, Hokkaido 060-0819, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan.
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
20
|
Daems M, Peacock HM, Jones EAV. Fluid flow as a driver of embryonic morphogenesis. Development 2020; 147:147/15/dev185579. [PMID: 32769200 DOI: 10.1242/dev.185579] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fluid flow is a powerful morphogenic force during embryonic development. The physical forces created by flowing fluids can either create morphogen gradients or be translated by mechanosensitive cells into biological changes in gene expression. In this Primer, we describe how fluid flow is created in different systems and highlight the important mechanosensitive signalling pathways involved for sensing and transducing flow during embryogenesis. Specifically, we describe how fluid flow helps establish left-right asymmetry in the early embryo and discuss the role of flow of blood, lymph and cerebrospinal fluid in sculpting the embryonic cardiovascular and nervous system.
Collapse
Affiliation(s)
- Margo Daems
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Hanna M Peacock
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
21
|
Fame RM, Lehtinen MK. Emergence and Developmental Roles of the Cerebrospinal Fluid System. Dev Cell 2020; 52:261-275. [PMID: 32049038 DOI: 10.1016/j.devcel.2020.01.027] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022]
Abstract
We summarize recent work illuminating how cerebrospinal fluid (CSF) regulates brain function. More than a protective fluid cushion and sink for waste, the CSF is an integral CNS component with dynamic and diverse roles emerging in parallel with the developing CNS. This review examines the current understanding about early CSF and its maturation and roles during CNS development and discusses open questions in the field. We focus on developmental changes in the ventricular system and CSF sources (including neural progenitors and choroid plexus). We also discuss concepts related to the development of fluid dynamics including flow, perivascular transport, drainage, and barriers.
Collapse
Affiliation(s)
- Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Development of Ependymal and Postnatal Neural Stem Cells and Their Origin from a Common Embryonic Progenitor. Cell Rep 2020; 27:429-441.e3. [PMID: 30970247 DOI: 10.1016/j.celrep.2019.01.088] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/05/2018] [Accepted: 01/23/2019] [Indexed: 11/20/2022] Open
Abstract
The adult mouse brain contains an extensive neurogenic niche in the lateral walls of the lateral ventricles. This epithelium, which has a unique pinwheel organization, contains multiciliated ependymal (E1) cells and neural stem cells (B1). This postnatal germinal epithelium develops from the embryonic ventricular zone, but the lineage relationship between E1 and B1 cells remains unknown. Distinct subpopulations of radial glia (RG) cells in late embryonic and early postnatal development either expand their apical domain >11-fold to form E1 cells or retain small apical domains that coalesce into the centers of pinwheels to form B1 cells. Using independent methods of lineage tracing, we show that individual RG cells can give rise to clones containing E1 and B1 cells. This study reveals key developmental steps in the formation of the postnatal germinal niche and the shared cellular origin of E1 and B1 cells.
Collapse
|
23
|
Derish I, Lee JKH, Wong-King-Cheong M, Babayeva S, Caplan J, Leung V, Shahinian C, Gravel M, Deans MR, Gros P, Torban E. Differential role of planar cell polarity gene Vangl2 in embryonic and adult mammalian kidneys. PLoS One 2020; 15:e0230586. [PMID: 32203543 PMCID: PMC7089571 DOI: 10.1371/journal.pone.0230586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
Planar cell polarity (PCP) pathway is crucial for tissue morphogenesis. Mutations in PCP genes cause multi-organ anomalies including dysplastic kidneys. Defective PCP signaling was postulated to contribute to cystogenesis in polycystic kidney disease. This work was undertaken to elucidate the role of the key PCP gene, Vangl2, in embryonic and postnatal renal tubules and ascertain whether its loss contributes to cyst formation and defective tubular function in mature animals. We generated mice with ubiquitous and collecting duct-restricted excision of Vangl2. We analyzed renal tubules in mutant and control mice at embryonic day E17.5 and postnatal days P1, P7, P30, P90, 6- and 9-month old animals. The collecting duct functions were analyzed in young and adult mutant and control mice. Loss of Vangl2 leads to profound tubular dilatation and microcysts in embryonic kidneys. Mechanistically, these abnormalities are caused by defective convergent extension (larger tubular cross-sectional area) and apical constriction (cuboidal cell shape and a reduction of activated actomyosin at the luminal surface). However, the embryonic tubule defects were rapidly resolved by Vangl2-independent mechanisms after birth. Normal collecting duct architecture and functions were found in young and mature animals. During embryogenesis, Vangl2 controls tubular size via convergent extension and apical constriction. However, rapidly after birth, PCP-dependent control of tubular size is switched to a PCP-independent regulatory mechanism. We conclude that loss of the Vangl2 gene is dispensable for tubular elongation and maintenance postnatally. It does not lead to cyst formation and is unlikely to contribute to polycystic kidney disease.
Collapse
Affiliation(s)
- Ida Derish
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Jeremy K. H. Lee
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Melanie Wong-King-Cheong
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Sima Babayeva
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Jillian Caplan
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Vicki Leung
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Chloe Shahinian
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Michel Gravel
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Michael R. Deans
- Division of Otolaryngology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Elena Torban
- Department of Medicine, McGill University and McGill University Health Center Research Institute, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
24
|
Fame RM, Cortés-Campos C, Sive HL. Brain Ventricular System and Cerebrospinal Fluid Development and Function: Light at the End of the Tube: A Primer with Latest Insights. Bioessays 2020; 42:e1900186. [PMID: 32078177 DOI: 10.1002/bies.201900186] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/02/2020] [Indexed: 12/12/2022]
Abstract
The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.
Collapse
Affiliation(s)
- Ryann M Fame
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | | | - Hazel L Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
25
|
Ringers C, Olstad EW, Jurisch-Yaksi N. The role of motile cilia in the development and physiology of the nervous system. Philos Trans R Soc Lond B Biol Sci 2019; 375:20190156. [PMID: 31884916 DOI: 10.1098/rstb.2019.0156] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Motile cilia are miniature, whip-like organelles whose beating generates a directional fluid flow. The flow generated by ciliated epithelia is a subject of great interest, as defective ciliary motility results in severe human diseases called motile ciliopathies. Despite the abundance of motile cilia in diverse organs including the nervous system, their role in organ development and homeostasis remains poorly understood. Recently, much progress has been made regarding the identity of motile ciliated cells and the role of motile-cilia-mediated flow in the development and physiology of the nervous system. In this review, we will discuss these recent advances from sensory organs, specifically the nose and the ear, to the spinal cord and brain ventricles. This article is part of the Theo Murphy meeting issue 'Unity and diversity of cilia in locomotion and transport'.
Collapse
Affiliation(s)
- Christa Ringers
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Emilie W Olstad
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Nathalie Jurisch-Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.,Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Edvard Griegs Gate 8, 7030 Trondheim, Norway
| |
Collapse
|
26
|
Mathewson AW, Berman DG, Moens CB. Microtubules are required for the maintenance of planar cell polarity in monociliated floorplate cells. Dev Biol 2019; 452:21-33. [PMID: 31029691 PMCID: PMC6661169 DOI: 10.1016/j.ydbio.2019.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 11/21/2022]
Abstract
The asymmetric localization of planar cell polarity (PCP) proteins is essential for the establishment of many planar polarized cellular processes, but the mechanisms that maintain these asymmetric distributions remain poorly understood. A body of evidence has tied oriented subapical microtubules (MTs) to the establishment of PCP protein polarity, yet recent studies have suggested that the MT cytoskeleton is later dispensable for the maintenance of this asymmetry. As MTs underlie the vesicular trafficking of membrane-bound proteins within cells, the requirement for MTs in the maintenance of PCP merited further investigation. We investigated the complex interactions between PCP proteins and the MT cytoskeleton in the polarized context of the floorplate of the zebrafish neural tube. We demonstrated that the progressive posterior polarization of the primary cilia of floorplate cells requires not only Vangl2 but also Fzd3a. We determined that GFP-Vangl2 asymmetrically localizes to anterior membranes whereas Fzd3a-GFP does not polarize on anterior or posterior membranes but maintains a cytosolic enrichment at the base of the primary cilium. Vesicular Fzd3a-GFP is rapidly trafficked along MTs primarily toward the apical membrane during a period of PCP maintenance, whereas vesicular GFP-Vangl2 is less frequently observed. Nocodazole-induced loss of MT polymerization disrupts basal body positioning as well as GFP-Vangl2 localization and reduces cytosolic Fzd3a-GFP movements. Removal of nocodazole after MT disruption restores MT polymerization but does not restore basal body polarity. Interestingly, GFP-Vangl2 repolarizes to anterior membranes and vesicular Fzd3a-GFP dynamics recover after multiple hours of recovery, even in the context of unpolarized basal bodies. Together our findings challenge previous work by revealing an ongoing role for MT-dependent transport of PCP proteins in maintaining both cellular and PCP protein asymmetry during development.
Collapse
Affiliation(s)
- Andrew W Mathewson
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | - Daniel G Berman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Biology Graduate Program, University of Washington, Seattle, WA, USA
| | - Cecilia B Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA; Biology Graduate Program, University of Washington, Seattle, WA, USA.
| |
Collapse
|
27
|
Winokurow N, Schumacher S. A role for polycystin-1 and polycystin-2 in neural progenitor cell differentiation. Cell Mol Life Sci 2019; 76:2851-2869. [PMID: 30895336 PMCID: PMC11105687 DOI: 10.1007/s00018-019-03072-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/17/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
Polycystin-1 (PC1) and polycystin-2 (PC2) are transmembrane proteins encoded by the Pkd1 and Pkd2 genes, respectively. Mutations in these genes are causative for the development of autosomal-dominant polycystic kidney disease. A prominent feature of this disease is an unbalanced cell proliferation. PC1 and PC2 physically interact to form a complex, which localizes to the primary cilia of renal epithelial cells. Recently, PC1 and PC2 have also been described to be present in primary cilia of radial glial cells (RGCs) and to contribute to the planar cell polarity of late RGCs and E1 ependymal cells. As neural progenitor cells (NPCs), early RGCs have to balance proliferation for expansion, or for self-renewal and differentiation to generate neurons. It is not known whether the polycystins play a role in this process. Here, we show that PC1 and PC2 are expressed in RGCs of the developing mouse cerebral cortex during neurogenesis. Loss-of-function analysis and cell-based assays reveal that a reduction of PC1 or PC2 expression leads to increased NPC proliferation, while the differentiation to neurons becomes impaired. The increased NPC proliferation is preceded by enhanced Notch signaling and accompanied by a rise in the number of symmetric cell divisions. The transcription factor STAT3 seems to be mechanistically important for polycystin signaling in NPCs as either STAT3 knockdown or inhibition of STAT3 function abrogates the increased proliferation driven by reduced polycystin expression. Our findings indicate that PC1 and PC2 are critical for maintaining a balance between proliferation and differentiation of NPCs.
Collapse
Affiliation(s)
- Natalie Winokurow
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
28
|
Abstract
For over a century, the centrosome has been an organelle more easily tracked than understood, and the study of its peregrinations within the cell remains a chief underpinning of its functional investigation. Increasing attention and new approaches have been brought to bear on mechanisms that control centrosome localization in the context of cleavage plane determination, ciliogenesis, directional migration, and immunological synapse formation, among other cellular and developmental processes. The Golgi complex, often linked with the centrosome, presents a contrasting case of a pleiomorphic organelle for which functional studies advanced somewhat more rapidly than positional tracking. However, Golgi orientation and distribution has emerged as an area of considerable interest with respect to polarized cellular function. This chapter will review our current understanding of the mechanism and significance of the positioning of these organelles.
Collapse
|
29
|
Saghaeian Jazi M, Samaei NM, Mowla SJ, Arefnezhad B, Kouhsar M. SOX2OT knockdown derived changes in mitotic regulatory gene network of cancer cells. Cancer Cell Int 2018; 18:129. [PMID: 30202240 PMCID: PMC6126007 DOI: 10.1186/s12935-018-0618-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 08/14/2018] [Indexed: 01/24/2023] Open
Abstract
Background SOX2 overlapping transcript (SOX2OT) is a long non-coding RNA, over-expressed in human tumor tissues and embryonic cells. Evidences support its function in the cell cycle; however there is no clear mechanism explaining its function in cell proliferation regulation. Here we investigated cancer cell response to SOX2OT knockdown by RNA sequencing. Methods SOX2OT expression was inhibited by siRNA in two cancer cell lines (A549, U-87 MG), then the RNA of treated cells were used for the cDNA library synthesis and RNA sequencing. The differentially expressed genes were used for functional enrichment and the gene expression network was analyzed to find the most relevant biological process with SOX2OT function. Furthermore, the expression change of candidate genes was measured by qRT-PCR for more confirmation and the cell cycle was monitored by PI staining. Results Our findings showed that SOX2OT knockdown affects the cellular gene expression generally with enriched cell proliferation and development biological process. Particularly, the cell cycle and mitotic regulatory genes expression including: CDK2, CDK2AP2, ACTR3, and chromosome structure associated genes like SMC4, INCENP and GNL3L are changed in treated cancer cells. Conclusion Our results propound SOX2OT association with cell cycle and mitosis regulation in cancer cells. Electronic supplementary material The online version of this article (10.1186/s12935-018-0618-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie Saghaeian Jazi
- 1Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nader Mansour Samaei
- 2Stem Cell Research Center, Golestan University of Medical Sciences, Po Box: 4934174611, Gorgan, Iran
| | - Seyed Javad Mowla
- 3Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Morteza Kouhsar
- 5Laboratory of System Biology and Bioinformatics (LBB), University of Tehran, Institute of Biochemistry and Biophysics, Tehran, Iran
| |
Collapse
|
30
|
Ohata S, Uga H, Okamoto H, Katada T. Small GTPase R-Ras participates in neural tube formation in zebrafish embryonic spinal cord. Biochem Biophys Res Commun 2018; 501:786-790. [PMID: 29772239 DOI: 10.1016/j.bbrc.2018.05.074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/12/2018] [Indexed: 10/16/2022]
Abstract
Ras related (R-Ras), a small GTPase, is involved in the maintenance of apico-basal polarity in neuroepithelial cells of the zebrafish hindbrain, axonal collapse in cultured murine hippocampal neurons, and maturation of blood vessels in adult mice. However, the role of R-Ras in neural tube formation remains unknown. Using antisense morpholino oligonucleotides (AMOs), we found that in the spinal cord of zebrafish embryos, the lumen was formed bilaterally in rras morphants, whereas it was formed at the midline in control embryos. As AMO can cause off-target effects, we generated rras mutant zebrafish lines using CRISPR/Cas9 technology. Although these rras mutant embryos did not have a bilateral lumen in the spinal cord, the following findings suggest that the phenotype is unlikely due to an off-target effect of rras AMO: 1) The rras morphant phenotype was rescued by an injection of AMO-resistant rras mRNA, and 2) a bilaterally segregated spinal cord was not observed in rras mutant embryos injected with rras AMO. The results suggest that the function of other ras family genes may be redundant in rras mutants. Previous research reported a bilaterally formed lumen in the spinal cord of zebrafish embryos with a mutation in a planar cell polarity (PCP) gene, van gogh-like 2 (vangl2). In the present study, in cultured cells, R-Ras was co-immunoprecipitated with Vangl2 but not with another PCP regulator, Pricke1. Interestingly, the interaction between R-Ras and Vangl2 was stronger in guanine-nucleotide free point mutants of R-Ras than in wild-type or constitutively active (GTP-bound) forms of R-Ras. R-Ras may regulate neural tube formation in cooperation with Vangl2 in the developing zebrafish spinal cord.
Collapse
Affiliation(s)
- Shinya Ohata
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan; RIKEN Center for Brain Science, Saitama, 351-0198, Japan.
| | - Hideko Uga
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Toshiaki Katada
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
31
|
Youn YH, Han YG. Primary Cilia in Brain Development and Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:11-22. [PMID: 29030052 PMCID: PMC5745523 DOI: 10.1016/j.ajpath.2017.08.031] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 08/02/2017] [Accepted: 08/17/2017] [Indexed: 01/20/2023]
Abstract
The primary cilium, a sensory appendage that is present in most mammalian cells, plays critical roles in signaling pathways and cell cycle progression. Mutations that affect the structure or function of primary cilia result in ciliopathies, a group of developmental and degenerative diseases that affect almost all organs and tissues. Our understanding of the constituents, development, and function of primary cilia has advanced considerably in recent years, revealing pathogenic mechanisms that potentially underlie ciliopathies. In the brain, the primary cilia are crucial for early patterning, neurogenesis, neuronal maturation and survival, and tumorigenesis, mostly through regulating cell cycle progression, Hedgehog signaling, and WNT signaling. We review these advances in our knowledge of primary cilia, focusing on brain development, and discuss the mechanisms that may underlie brain abnormalities in ciliopathies.
Collapse
Affiliation(s)
- Yong Ha Youn
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Young-Goo Han
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
32
|
The mouse Jhy gene regulates ependymal cell differentiation and ciliogenesis. PLoS One 2017; 12:e0184957. [PMID: 29211732 PMCID: PMC5718522 DOI: 10.1371/journal.pone.0184957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
During the first postnatal week of mouse development, radial glial cells lining the ventricles of the brain differentiate into ependymal cells, undergoing a morphological change from pseudostratified cuboidal cells to a flattened monolayer. Concomitant with this change, multiple motile cilia are generated and aligned on each nascent ependymal cell. Proper ependymal cell development is crucial to forming the brain tissue:CSF barrier, and to the establishment of ciliary CSF flow, but the mechanisms that regulate this differentiation event are poorly understood. The JhylacZ mouse line carries an insertional mutation in the Jhy gene (formerly 4931429I11Rik), and homozygous JhylacZ/lacZ mice develop a rapidly progressive juvenile hydrocephalus, with defects in ependymal cilia morphology and ultrastructure. Here we show that beyond just defective motile cilia, JhylacZ/lacZ mice display abnormal ependymal cell differentiation. Ventricular ependyma in JhylacZ/lacZ mice retain an unorganized and multi-layered morphology, representative of undifferentiated ependymal (radial glial) cells, and they show altered expression of differentiation markers. Most JhylacZ/lacZ ependymal cells do eventually acquire some differentiated ependymal characteristics, suggesting a delay, rather than a block, in the differentiation process, but ciliogenesis remains perturbed. JhylacZ/lacZ ependymal cells also manifest disruptions in adherens junction formation, with altered N-cadherin localization, and have defects in the polarized organization of the apical motile cilia that do form. Functional studies showed that cilia of JhylacZ/lacZ mice have severely reduced motility, a potential cause for the development of hydrocephalus. This work shows that JHY does not only control ciliogenesis, but is a crucial component of the ependymal differentiation process, with ciliary defects likely a consequence of altered ependymal differentiation.
Collapse
|
33
|
CSF in the ventricles of the brain behaves as a relay medium for arteriovenous pulse wave phase coupling. PLoS One 2017; 12:e0181025. [PMID: 29141045 PMCID: PMC5687699 DOI: 10.1371/journal.pone.0181025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/21/2017] [Indexed: 11/19/2022] Open
Abstract
The ventricles of the brain remain perhaps the largest anatomic structure in the human body without established primary purpose, even though their existence has been known at least since described by Aristotle. We hypothesize that the ventricles help match a stroke volume of arterial blood that arrives into the rigid cranium with an equivalent volume of ejected venous blood by spatially configuring cerebrospinal fluid (CSF) to act as a low viscosity relay medium for arteriovenous pulse wave (PW) phase coupling. We probe the hypothesis by comparing the spatiotemporal behavior of vascular PW about the ventricular surfaces in piglets to internal observations of ventricle wall motions and adjacent CSF pressure variations in humans. With wavelet brain angiography data obtained from piglets, we map the travel relative to brain pulse motion of arterial and venous PWs over the ventricle surfaces. We find that arterial PWs differ in CF phase from venous PWs over the surfaces of the ventricles consistent with arteriovenous PW phase coupling. We find a spatiotemporal difference in vascular PW phase between the ventral and dorsal ventricular surfaces, with the PWs arriving slightly sooner to the ventral surfaces. In humans undergoing neuroendoscopic surgery for hydrocephalus, we measure directly ventricle wall motions and the adjacent internal CSF pressure variations. We find that CSF pressure peaks slightly earlier in the ventral Third Ventricle than the dorsal Lateral Ventricle. When matched anatomically, the peri-ventricular vascular PW phase distribution in piglets complements the endo-ventricular CSF PW phase distribution in humans. This is consistent with a role for the ventricles in arteriovenous PW coupling and may add a framework for understanding hydrocephalus and other disturbances of intracranial pressure.
Collapse
|
34
|
Omran AJA, Saternos HC, Althobaiti YS, Wisner A, Sari Y, Nauli SM, AbouAlaiwi WA. Alcohol consumption impairs the ependymal cilia motility in the brain ventricles. Sci Rep 2017; 7:13652. [PMID: 29057897 PMCID: PMC5651853 DOI: 10.1038/s41598-017-13947-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/02/2017] [Indexed: 12/13/2022] Open
Abstract
Ependymal cilia protrude into the central canal of the brain ventricles and spinal cord to circulate the cerebral spinal fluid (CSF). Ependymal cilia dysfunction can hinder the movement of CSF leading to an abnormal accumulation of CSF within the brain known as hydrocephalus. Although the etiology of hydrocephalus was studied before, the effects of ethanol ingestion on ependymal cilia function have not been investigated in vivo. Here, we report three distinct types of ependymal cilia, type-I, type-II and type-III classified based upon their beating frequency, their beating angle, and their distinct localization within the mouse brain-lateral ventricle. Our studies show for the first time that oral gavage of ethanol decreased the beating frequency of all three types of ependymal cilia in both the third and the lateral rat brain ventricles in vivo. Furthermore, we show for the first time that hydin, a hydrocephalus-inducing gene product whose mutation impairs ciliary motility, and polycystin-2, whose ablation is associated with hydrocephalus are colocalized to the ependymal cilia. Thus, our studies reinforce the presence of three types of ependymal cilia in the brain ventricles and demonstrate the involvement of ethanol as a risk factor for the impairment of ependymal cilia motility in the brain.
Collapse
Affiliation(s)
- Alzahra J Al Omran
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, Ohio, USA
| | - Hannah C Saternos
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, Ohio, USA
| | - Yusuf S Althobaiti
- Taif University, College of Pharmacy, Department of Pharmacology and Toxicology, Taif, Kingdom of Saudi Arabia
| | - Alexander Wisner
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, Ohio, USA
| | - Youssef Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, Ohio, USA
| | - Surya M Nauli
- Chapman University, College of Pharmacy, Irvine, California, USA
| | - Wissam A AbouAlaiwi
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, Toledo, Ohio, USA.
| |
Collapse
|
35
|
Daple Coordinates Planar Polarized Microtubule Dynamics in Ependymal Cells and Contributes to Hydrocephalus. Cell Rep 2017; 20:960-972. [DOI: 10.1016/j.celrep.2017.06.089] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 05/22/2017] [Accepted: 06/29/2017] [Indexed: 11/15/2022] Open
|
36
|
Abstract
Multiciliated cells are epithelial cells that are in contact with bodily fluids and are required for the proper function of major organs including the brain, the respiratory system and the reproductive tracts. Their multiple motile cilia beat unidirectionally to remove particles of external origin from their surface and/or drive cells or fluids into the lumen of the organs. Multiciliated cells in the brain are produced once, almost exclusively during embryonic development, whereas in respiratory tracts and oviducts they regenerate throughout life. In this Review, we provide a cell-to-organ overview of multiciliated cells and highlight recent studies that have greatly increased our understanding of the mechanisms driving the development and function of these cells in vertebrates. We discuss cell fate determination and differentiation of multiciliated cells, and provide a comprehensive account of their locations and functions in mammals.
Collapse
|
37
|
Ferreira RR, Vermot J. The balancing roles of mechanical forces during left-right patterning and asymmetric morphogenesis. Mech Dev 2017; 144:71-80. [DOI: 10.1016/j.mod.2016.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022]
|
38
|
Mirzadeh Z, Kusne Y, Duran-Moreno M, Cabrales E, Gil-Perotin S, Ortiz C, Chen B, Garcia-Verdugo JM, Sanai N, Alvarez-Buylla A. Bi- and uniciliated ependymal cells define continuous floor-plate-derived tanycytic territories. Nat Commun 2017; 8:13759. [PMID: 28067220 PMCID: PMC5477523 DOI: 10.1038/ncomms13759] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023] Open
Abstract
Multiciliated ependymal (E1) cells line the brain ventricles and are essential for brain homeostasis. We previously identified in the lateral ventricles a rare ependymal subpopulation (E2) with only two cilia and unique basal bodies. Here we show that E2 cells form a distinct biciliated epithelium extending along the ventral third into the fourth ventricle. In the third ventricle floor, apical profiles with only primary cilia define an additional uniciliated (E3) epithelium. E2 and E3 cells' ultrastructure, marker expression and basal processes indicate that they correspond to subtypes of tanycytes. Using sonic hedgehog lineage tracing, we show that the third and fourth ventricle E2 and E3 epithelia originate from the anterior floor plate. E2 and E3 cells complete their differentiation 2-3 weeks after birth, suggesting a link to postnatal maturation. These data reveal discrete bands of E2 and E3 cells that may relay information from the CSF to underlying neural circuits along the ventral midline.
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Division of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA
| | - Yael Kusne
- Division of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA
| | - Maria Duran-Moreno
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, CIBERNED, Universidad de Valencia, Valencia 46980, Spain
| | - Elaine Cabrales
- Division of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA
| | - Sara Gil-Perotin
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, CIBERNED, Universidad de Valencia, Valencia 46980, Spain
| | - Christian Ortiz
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California 95064, USA
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California 95064, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, CIBERNED, Universidad de Valencia, Valencia 46980, Spain
| | - Nader Sanai
- Division of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery and The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, 35 Medical Center Way, Room RMB-1036, Campus Box 0525, San Francisco, California 94143, USA
| |
Collapse
|
39
|
Vilboux T, Malicdan MCV, Roney JC, Cullinane AR, Stephen J, Yildirimli D, Bryant J, Fischer R, Vemulapalli M, Mullikin JC, Steinbach PJ, Gahl WA, Gunay-Aygun M. CELSR2, encoding a planar cell polarity protein, is a putative gene in Joubert syndrome with cortical heterotopia, microophthalmia, and growth hormone deficiency. Am J Med Genet A 2017; 173:661-666. [PMID: 28052552 DOI: 10.1002/ajmg.a.38005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/19/2016] [Indexed: 11/07/2022]
Abstract
Joubert syndrome is a ciliopathy characterized by a specific constellation of central nervous system malformations that result in the pathognomonic "molar tooth sign" on imaging. More than 27 genes are associated with Joubert syndrome, but some patients do not have mutations in any of these genes. Celsr1, Celsr2, and Celsr3 are the mammalian orthologues of the drosophila planar cell polarity protein, flamingo; they play important roles in neural development, including axon guidance, neuronal migration, and cilium polarity. Here, we report bi-allelic mutations in CELSR2 in a Joubert patient with cortical heterotopia, microophthalmia, and growth hormone deficiency. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Thierry Vilboux
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
- Inova Translational Medicine Institute, Falls Church, Virginia
| | - May Christine V Malicdan
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Joseph C Roney
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrew R Cullinane
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
- Department of Anatomy, Howard University College of Medicine, Washington DC
| | - Joshi Stephen
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Deniz Yildirimli
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Joy Bryant
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Roxanne Fischer
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Meghana Vemulapalli
- NIH Intramural Sequencing Center (NISC), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - James C Mullikin
- NIH Intramural Sequencing Center (NISC), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter J Steinbach
- Center for Molecular Modeling, Center for Information Technology, National Institutes of Health, Bethesda, Maryland
| | - William A Gahl
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Meral Gunay-Aygun
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Foerster P, Daclin M, Asm S, Faucourt M, Boletta A, Genovesio A, Spassky N. mTORC1 signaling and primary cilia are required for brain ventricle morphogenesis. Development 2016; 144:201-210. [PMID: 27993979 PMCID: PMC5394754 DOI: 10.1242/dev.138271] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 12/05/2016] [Indexed: 01/06/2023]
Abstract
Radial glial cells (RCGs) are self-renewing progenitor cells that give rise to neurons and glia during embryonic development. Throughout neurogenesis, these cells contact the cerebral ventricles and bear a primary cilium. Although the role of the primary cilium in embryonic patterning has been studied, its role in brain ventricular morphogenesis is poorly characterized. Using conditional mutants, we show that the primary cilia of radial glia determine the size of the surface of their ventricular apical domain through regulation of the mTORC1 pathway. In cilium-less mutants, the orientation of the mitotic spindle in radial glia is also significantly perturbed and associated with an increased number of basal progenitors. The enlarged apical domain of RGCs leads to dilatation of the brain ventricles during late embryonic stages (ventriculomegaly), which initiates hydrocephalus during postnatal stages. These phenotypes can all be significantly rescued by treatment with the mTORC1 inhibitor rapamycin. These results suggest that primary cilia regulate ventricle morphogenesis by acting as a brake on the mTORC1 pathway. This opens new avenues for the diagnosis and treatment of hydrocephalus.
Collapse
Affiliation(s)
- Philippe Foerster
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| | - Marie Daclin
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| | - Shihavuddin Asm
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| | - Marion Faucourt
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Auguste Genovesio
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| | - Nathalie Spassky
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), INSERM U1024, and CNRS UMR 8197, PSL Research University, 46 rue d'Ulm, Paris 75005, France
| |
Collapse
|
41
|
Abstract
Many animal cells assemble single cilia involved in motile and/or sensory functions. In contrast, multiciliated cells (MCCs) assemble up to 300 motile cilia that beat in a coordinate fashion to generate a directional fluid flow. In the human airways, the brain, and the oviduct, MCCs allow mucus clearance, cerebrospinal fluid circulation, and egg transportation, respectively. Impairment of MCC function leads to chronic respiratory infections and increased risks of hydrocephalus and female infertility. MCC differentiation during development or repair involves the activation of a regulatory cascade triggered by the inhibition of Notch activity in MCC progenitors. The downstream events include the simultaneous assembly of a large number of basal bodies (BBs)-from which cilia are nucleated-in the cytoplasm of the differentiating MCCs, their migration and docking at the plasma membrane associated to an important remodeling of the actin cytoskeleton, and the assembly and polarization of motile cilia. The direction of ciliary beating is coordinated both within cells and at the tissue level by a combination of planar polarity cues affecting BB position and hydrodynamic forces that are both generated and sensed by the cilia. Herein, we review the mechanisms controlling the specification and differentiation of MCCs and BB assembly and organization at the apical surface, as well as ciliary assembly and coordination in MCCs.
Collapse
Affiliation(s)
- Alice Meunier
- Institut de Biologie de l'Ecole Normale Supérieure, Institut National de la Santé et de la Recherche Médicale U1024, Centre National de la Recherche Scientifique UMR8197, 75005 Paris, France
| | - Juliette Azimzadeh
- Institut Jacques Monod, Centre National de la Recherche Scientifique UMR7592, Université Paris-Diderot, 75013 Paris, France
| |
Collapse
|
42
|
Campos Y, Qiu X, Gomero E, Wakefield R, Horner L, Brutkowski W, Han YG, Solecki D, Frase S, Bongiovanni A, d'Azzo A. Alix-mediated assembly of the actomyosin-tight junction polarity complex preserves epithelial polarity and epithelial barrier. Nat Commun 2016; 7:11876. [PMID: 27336173 PMCID: PMC4931029 DOI: 10.1038/ncomms11876] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Maintenance of epithelial cell polarity and epithelial barrier relies on the spatial organization of the actin cytoskeleton and proper positioning/assembly of intercellular junctions. However, how these processes are regulated is poorly understood. Here we reveal a key role for the multifunctional protein Alix in both processes. In a knockout mouse model of Alix, we identified overt structural changes in the epithelium of the choroid plexus and in the ependyma, such as asymmetrical cell shape and size, misplacement and abnormal beating of cilia, blebbing of the microvilli. These defects culminate in excessive cell extrusion, enlargement of the lateral ventricles and hydrocephalus. Mechanistically, we find that by interacting with F-actin, the Par complex and ZO-1, Alix ensures the formation and maintenance of the apically restricted actomyosin-tight junction complex. We propose that in this capacity Alix plays a role in the establishment of apical-basal polarity and in the maintenance of the epithelial barrier.
Collapse
Affiliation(s)
- Yvan Campos
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Xiaohui Qiu
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Elida Gomero
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Randall Wakefield
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Linda Horner
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Wojciech Brutkowski
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Young-Goo Han
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - David Solecki
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Sharon Frase
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology, National Research Council, 90146 Palermo, Italy
| | - Alessandra d'Azzo
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| |
Collapse
|
43
|
Fame RM, Chang JT, Hong A, Aponte-Santiago NA, Sive H. Directional cerebrospinal fluid movement between brain ventricles in larval zebrafish. Fluids Barriers CNS 2016; 13:11. [PMID: 27329482 PMCID: PMC4915066 DOI: 10.1186/s12987-016-0036-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/09/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) contained within the brain ventricles contacts neuroepithelial progenitor cells during brain development. Dynamic properties of CSF movement may limit locally produced factors to specific regions of the developing brain. However, there is no study of in vivo CSF dynamics between ventricles in the embryonic brain. We address CSF movement using the zebrafish larva, during the major period of developmental neurogenesis. METHODS CSF movement was monitored at two stages of zebrafish development: early larva [pharyngula stage; 27-30 h post-fertilization (hpf)] and late larva (hatching period; 51-54 hpf) using photoactivatable Kaede protein to calculate average maximum CSF velocity between ventricles. Potential roles for heartbeat in early CSF movement were investigated using tnnt2a mutant fish (tnnt2a (-/-)) and chemical [2,3 butanedione monoxime (BDM)] treatment. Cilia motility was monitored at these stages using the Tg(βact:Arl13b-GFP) transgenic fish line. RESULTS In wild-type early larva there is net CSF movement from the telencephalon to the combined diencephalic/mesencephalic superventricle. This movement directionality reverses at late larval stage. CSF moves directionally from diencephalic to rhombencephalic ventricles at both stages examined, with minimal movement from rhombencephalon to diencephalon. Directional movement is partially dependent on heartbeat, as indicated in assays of tnnt2a (-/-) fish and after BDM treatment. Brain cilia are immotile at the early larval stage. CONCLUSION These data demonstrate directional movement of the embryonic CSF in the zebrafish model during the major period of developmental neurogenesis. A key conclusion is that CSF moves preferentially from the diencephalic into the rhombencephalic ventricle. In addition, the direction of CSF movement between telencephalic and diencephalic ventricles reverses between the early and late larval stages. CSF movement is partially dependent on heartbeat. At early larval stage, the absence of motile cilia indicates that cilia likely do not direct CSF movement. These data suggest that CSF components may be compartmentalized and could contribute to specialization of the early brain. In addition, CSF movement may also provide directional mechanical signaling.
Collapse
Affiliation(s)
- Ryann M Fame
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA, 02142, USA
| | - Jessica T Chang
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA, 02142, USA.,Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139-4307, USA
| | - Alex Hong
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139-4307, USA
| | | | - Hazel Sive
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA, 02142, USA. .,Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139-4307, USA.
| |
Collapse
|
44
|
Ohata S, Alvarez-Buylla A. Planar Organization of Multiciliated Ependymal (E1) Cells in the Brain Ventricular Epithelium. Trends Neurosci 2016; 39:543-551. [PMID: 27311928 DOI: 10.1016/j.tins.2016.05.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 01/17/2023]
Abstract
Cerebrospinal fluid (CSF) continuously flows through the cerebral ventricles, a process essential for brain homeostasis. Multiciliated ependymal (E1) cells line the walls of the ventricles and contribute importantly to CSF flow through ciliary beating. Key to this function is the rotational and translational planar cell polarity (PCP) of E1 cells. Defects in the PCP of E1 cells can result in abnormal CSF accumulation and hydrocephalus. Here, we integrate recent data on the roles of early CSF flow in the embryonic ventricles, PCP regulators (e.g., Vangl2 and Dishevelled), and cytoskeletal networks in the establishment, refinement, and maintenance of E1 cells' PCP. The planar organization mechanisms of E1 cells could explain how CSF flow contributes to brain function and may help in the diagnosis and prevention of hydrocephalus.
Collapse
Affiliation(s)
- Shinya Ohata
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan.
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
45
|
Kim S, Nie H, Nesin V, Tran U, Outeda P, Bai CX, Keeling J, Maskey D, Watnick T, Wessely O, Tsiokas L. The polycystin complex mediates Wnt/Ca(2+) signalling. Nat Cell Biol 2016; 18:752-764. [PMID: 27214281 PMCID: PMC4925210 DOI: 10.1038/ncb3363] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/22/2016] [Indexed: 01/22/2023]
Abstract
WNT ligands induce Ca2+ signaling on target cells. PKD1 (Polycystin 1) is considered an orphan, atypical G protein coupled receptor complexed with TRPP2 (Polycystin 2 or PKD2), a Ca2+-permeable ion channel. Inactivating mutations in their genes cause autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases. Here, we show that WNTs bind to the extracellular domain of PKD1 and induce whole cell currents and Ca2+ influx dependent on TRPP2. Pathogenic PKD1 or PKD2 mutations that abrogate complex formation, compromise cell surface expression of PKD1, or reduce TRPP2 channel activity suppress activation by WNTs. Pkd2−/− fibroblasts lack WNT-induced Ca2+ currents and are unable to polarize during directed cell migration. In Xenopus embryos, PKD1, Dishevelled 2 (DVL2), and WNT9A act within the same pathway to preserve normal tubulogenesis. These data define PKD1 as a WNT (co)receptor and implicate defective WNT/Ca2+ signaling as one of the causes of ADPKD.
Collapse
Affiliation(s)
- Seokho Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Hongguang Nie
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA.,Institute of Metabolic Disease Research and Drug Development, China Medical University, Liaoning Shenyang, 110001 China (H.N)
| | - Vasyl Nesin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Uyen Tran
- Department of Cellular and Molecular Medicine, Cleveland Clinic, 9500 Euclid Avenue/NC10, Cleveland, OH 44195, USA
| | - Patricia Outeda
- Division of Nephrology, Baltimore PKD Research and Clinical Core Center, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Chang-Xi Bai
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA.,Department of Advanced Research on Mongolian Medicine, Research Institute for Mongolian Medicine, Inner Mongolia Medical University, Hohhot 010110, Inner Mongolia, China (CB)
| | - Jacob Keeling
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Dipak Maskey
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| | - Terry Watnick
- Division of Nephrology, Baltimore PKD Research and Clinical Core Center, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Oliver Wessely
- Department of Cellular and Molecular Medicine, Cleveland Clinic, 9500 Euclid Avenue/NC10, Cleveland, OH 44195, USA
| | - Leonidas Tsiokas
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
46
|
Pkd Proteins Team Up to Tell Cilia Which Way to Go. J Neurosci 2016; 36:643-5. [PMID: 26791196 DOI: 10.1523/jneurosci.3684-15.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
47
|
Hirota Y, Sawada M, Huang SH, Ogino T, Ohata S, Kubo A, Sawamoto K. Roles of Wnt Signaling in the Neurogenic Niche of the Adult Mouse Ventricular-Subventricular Zone. Neurochem Res 2015; 41:222-30. [PMID: 26572545 DOI: 10.1007/s11064-015-1766-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
Abstract
In many animal species, the production of new neurons (neurogenesis) occurs throughout life, in a specialized germinal region called the ventricular-subventricular zone (V-SVZ). In this region, neural stem cells undergo self-renewal and generate neural progenitor cells and new neurons. In the olfactory system, the new neurons migrate rostrally toward the olfactory bulb, where they differentiate into mature interneurons. V-SVZ-derived new neurons can also migrate toward sites of brain injury, where they contribute to neural regeneration. Recent studies indicate that two major branches of the Wnt signaling pathway, the Wnt/β-catenin and Wnt/planar cell polarity pathways, play essential roles in various facets of adult neurogenesis. Here, we review the Wnt signaling-mediated regulation of adult neurogenesis in the V-SVZ under physiological and pathological conditions.
Collapse
Affiliation(s)
- Yuki Hirota
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Shih-Hui Huang
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Takashi Ogino
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Shinya Ohata
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Akiharu Kubo
- Department of Dermatology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.
| |
Collapse
|
48
|
Nigro EA, Castelli M, Boletta A. Role of the Polycystins in Cell Migration, Polarity, and Tissue Morphogenesis. Cells 2015; 4:687-705. [PMID: 26529018 PMCID: PMC4695853 DOI: 10.3390/cells4040687] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022] Open
Abstract
Cystic kidney diseases (CKD) is a class of disorders characterized by ciliary dysfunction and, therefore, belonging to the ciliopathies. The prototype CKD is autosomal dominant polycystic kidney disease (ADPKD), whose mutated genes encode for two membrane-bound proteins, polycystin-1 (PC-1) and polycystin-2 (PC-2), of unknown function. Recent studies on CKD-associated genes identified new mechanisms of morphogenesis that are central for establishment and maintenance of proper renal tubular diameter. During embryonic development in the mouse and lower vertebrates a convergent-extension (CE)-like mechanism based on planar cell polarity (PCP) and cellular intercalation is involved in “sculpting” the tubules into a narrow and elongated shape. Once the appropriate diameter is established, further elongation occurs through oriented cell division (OCD). The polycystins (PCs) regulate some of these essential processes. In this review we summarize recent work on the role of PCs in regulating cell migration, the cytoskeleton, and front-rear polarity. These important properties are essential for proper morphogenesis of the renal tubules and the lymphatic vessels. We highlight here several open questions and controversies. Finally, we try to outline some of the next steps required to study these processes and their relevance in physiological and pathological conditions.
Collapse
Affiliation(s)
- Elisa Agnese Nigro
- Division of Genetics and Cell Biology, Dibit, IRCCS-San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy.
| | - Maddalena Castelli
- Division of Genetics and Cell Biology, Dibit, IRCCS-San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy.
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, Dibit, IRCCS-San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|