1
|
Niklaus S, Glasauer SMK, Kovermann P, Farshori KF, Cadetti L, Früh S, Rieser NN, Gesemann M, Zang J, Fahlke C, Neuhauss SCF. Glutamate transporters are involved in direct inhibitory synaptic transmission in the vertebrate retina. Open Biol 2024; 14:240140. [PMID: 39079673 PMCID: PMC11288666 DOI: 10.1098/rsob.240140] [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: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 08/03/2024] Open
Abstract
In the central nervous system of vertebrates, glutamate serves as the primary excitatory neurotransmitter. However, in the retina, glutamate released from photoreceptors causes hyperpolarization in post-synaptic ON-bipolar cells through a glutamate-gated chloride current, which seems paradoxical. Our research reveals that this current is modulated by two excitatory glutamate transporters, EAAT5b and EAAT7. In the zebrafish retina, these transporters are located at the dendritic tips of ON-bipolar cells and interact with all four types of cone photoreceptors. The absence of these transporters leads to a decrease in ON-bipolar cell responses, with eaat5b mutants being less severely affected than eaat5b/eaat7 double mutants, which also exhibit altered response kinetics. Biophysical investigations establish that EAAT7 is an active glutamate transporter with a predominant anion conductance. Our study is the first to demonstrate the direct involvement of post-synaptic glutamate transporters in inhibitory direct synaptic transmission at a central nervous system synapse.
Collapse
Affiliation(s)
- Stephanie Niklaus
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Stella M. K. Glasauer
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Peter Kovermann
- Institute of Biological Information Processing, Molekular- und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Leo-Brandt-Strasse, 52425 Jülich, Germany
| | - Kulsum F. Farshori
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Lucia Cadetti
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Simon Früh
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nicolas N. Rieser
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Matthias Gesemann
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jingjing Zang
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Christoph Fahlke
- Institute of Biological Information Processing, Molekular- und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Leo-Brandt-Strasse, 52425 Jülich, Germany
| | - Stephan C. F. Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
2
|
Passchier EMJ, Bisseling Q, Helman G, van Spaendonk RML, Simons C, Olsthoorn RCL, van der Veen H, Abbink TEM, van der Knaap MS, Min R. Megalencephalic leukoencephalopathy with subcortical cysts: a variant update and review of the literature. Front Genet 2024; 15:1352947. [PMID: 38487253 PMCID: PMC10938252 DOI: 10.3389/fgene.2024.1352947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024] Open
Abstract
The leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC) is characterized by infantile-onset macrocephaly and chronic edema of the brain white matter. With delayed onset, patients typically experience motor problems, epilepsy and slow cognitive decline. No treatment is available. Classic MLC is caused by bi-allelic recessive pathogenic variants in MLC1 or GLIALCAM (also called HEPACAM). Heterozygous dominant pathogenic variants in GLIALCAM lead to remitting MLC, where patients show a similar phenotype in early life, followed by normalization of white matter edema and no clinical regression. Rare patients with heterozygous dominant variants in GPRC5B and classic MLC were recently described. In addition, two siblings with bi-allelic recessive variants in AQP4 and remitting MLC have been identified. The last systematic overview of variants linked to MLC dates back to 2006. We provide an updated overview of published and novel variants. We report on genetic variants from 508 patients with MLC as confirmed by MRI diagnosis (258 from our database and 250 extracted from 64 published reports). We describe 151 unique MLC1 variants, 29 GLIALCAM variants, 2 GPRC5B variants and 1 AQP4 variant observed in these MLC patients. We include experiments confirming pathogenicity for some variants, discuss particularly notable variants, and provide an overview of recent scientific and clinical insight in the pathophysiology of MLC.
Collapse
Affiliation(s)
- Emma M. J. Passchier
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Quinty Bisseling
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Guy Helman
- Translational Bioinformatics, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
| | | | - Cas Simons
- Translational Bioinformatics, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | | | - Hieke van der Veen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Truus E. M. Abbink
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
3
|
Stogsdill JA, Harwell CC, Goldman SA. Astrocytes as master modulators of neural networks: Synaptic functions and disease-associated dysfunction of astrocytes. Ann N Y Acad Sci 2023; 1525:41-60. [PMID: 37219367 DOI: 10.1111/nyas.15004] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Astrocytes are the most abundant glial cell type in the central nervous system and are essential to the development, plasticity, and maintenance of neural circuits. Astrocytes are heterogeneous, with their diversity rooted in developmental programs modulated by the local brain environment. Astrocytes play integral roles in regulating and coordinating neural activity extending far beyond their metabolic support of neurons and other brain cell phenotypes. Both gray and white matter astrocytes occupy critical functional niches capable of modulating brain physiology on time scales slower than synaptic activity but faster than those adaptive responses requiring a structural change or adaptive myelination. Given their many associations and functional roles, it is not surprising that astrocytic dysfunction has been causally implicated in a broad set of neurodegenerative and neuropsychiatric disorders. In this review, we focus on recent discoveries concerning the contributions of astrocytes to the function of neural networks, with a dual focus on the contribution of astrocytes to synaptic development and maturation, and on their role in supporting myelin integrity, and hence conduction and its regulation. We then address the emerging roles of astrocytic dysfunction in disease pathogenesis and on potential strategies for targeting these cells for therapeutic purposes.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Steven A Goldman
- Sana Biotechnology Inc., Cambridge, Massachusetts, USA
- Center for Translational Neuromedicine, University of Rochester, Rochester, New York, USA
- University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
4
|
Stemerdink M, Broekman S, Peters T, Kremer H, de Vrieze E, van Wijk E. Generation and Characterization of a Zebrafish Model for ADGRV1-Associated Retinal Dysfunction Using CRISPR/Cas9 Genome Editing Technology. Cells 2023; 12:1598. [PMID: 37371069 DOI: 10.3390/cells12121598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Worldwide, around 40,000 people progressively lose their eyesight as a consequence of retinitis pigmentosa (RP) caused by pathogenic variants in the ADGRV1 gene, for which currently no treatment options exist. A model organism that mimics the human phenotype is essential to unravel the exact pathophysiological mechanism underlying ADGRV1-associated RP, and to evaluate future therapeutic strategies. The introduction of CRISPR/Cas-based genome editing technologies significantly improved the possibilities of generating mutant models in a time- and cost-effective manner. Zebrafish have been recognized as a suitable model to study Usher syndrome-associated retinal dysfunction. Using CRISPR/Cas9 technology we introduced a 4bp deletion in adgrv1 exon 9 (adgrv1rmc22). Immunohistochemical analysis showed that Adgrv1 was absent from the region of the photoreceptor connecting cilium in the adgrv1rmc22 zebrafish retina. Here, the absence of Adgrv1 also resulted in reduced levels of the USH2 complex members usherin and Whrnb, suggesting that Adgrv1 interacts with usherin and Whrnb in zebrafish photoreceptors. When comparing adgrv1rmc22 zebrafish with wild-type controls, we furthermore observed increased levels of aberrantly localized rhodopsin in the photoreceptor cell body, and decreased electroretinogram (ERG) B-wave amplitudes which indicate that the absence of Adgrv1 results in impaired retinal function. Based on these findings we present the adgrv1rmc22 zebrafish as the first ADGRV1 mutant model that displays an early retinal dysfunction. Moreover, the observed phenotypic changes can be used as quantifiable outcome measures when evaluating the efficacy of future novel therapeutic strategies for ADGRV1-associated RP.
Collapse
Affiliation(s)
- Merel Stemerdink
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Sanne Broekman
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Theo Peters
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Hannie Kremer
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Erwin van Wijk
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
5
|
Nowacki JC, Fields AM, Fu MM. Emerging cellular themes in leukodystrophies. Front Cell Dev Biol 2022; 10:902261. [PMID: 36003149 PMCID: PMC9393611 DOI: 10.3389/fcell.2022.902261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Leukodystrophies are a broad spectrum of neurological disorders that are characterized primarily by deficiencies in myelin formation. Clinical manifestations of leukodystrophies usually appear during childhood and common symptoms include lack of motor coordination, difficulty with or loss of ambulation, issues with vision and/or hearing, cognitive decline, regression in speech skills, and even seizures. Many cases of leukodystrophy can be attributed to genetic mutations, but they have diverse inheritance patterns (e.g., autosomal recessive, autosomal dominant, or X-linked) and some arise from de novo mutations. In this review, we provide an updated overview of 35 types of leukodystrophies and focus on cellular mechanisms that may underlie these disorders. We find common themes in specialized functions in oligodendrocytes, which are specialized producers of membranes and myelin lipids. These mechanisms include myelin protein defects, lipid processing and peroxisome dysfunction, transcriptional and translational dysregulation, disruptions in cytoskeletal organization, and cell junction defects. In addition, non-cell-autonomous factors in astrocytes and microglia, such as autoimmune reactivity, and intercellular communication, may also play a role in leukodystrophy onset. We hope that highlighting these themes in cellular dysfunction in leukodystrophies may yield conceptual insights on future therapeutic approaches.
Collapse
|
6
|
GPR37 Receptors and Megalencephalic Leukoencephalopathy with Subcortical Cysts. Int J Mol Sci 2022; 23:ijms23105528. [PMID: 35628339 PMCID: PMC9144339 DOI: 10.3390/ijms23105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of vacuolating leukodystrophy (white matter disorder), which is mainly caused by defects in MLC1 or glial cell adhesion molecule (GlialCAM) proteins. In addition, autoantibodies to GlialCAM are involved in the pathology of multiple sclerosis. MLC1 and GLIALCAM genes encode for membrane proteins of unknown function, which has been linked to the regulation of different ion channels and transporters, such as the chloride channel VRAC (volume regulated anion channel), ClC-2 (chloride channel 2), and connexin 43 or the Na+/K+-ATPase pump. However, the mechanisms by which MLC proteins regulate these ion channels and transporters, as well as the exact function of MLC proteins remain obscure. It has been suggested that MLC proteins might regulate signalling pathways, but the mechanisms involved are, at present, unknown. With the aim of answering these questions, we have recently described the brain GlialCAM interactome. Within the identified proteins, we could validate the interaction with several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptors GPR37L1 and GPR37. In this review, we summarize new aspects of the pathophysiology of MLC disease and key aspects of the interaction between GPR37 receptors and MLC proteins.
Collapse
|
7
|
Lanciotti A, Brignone MS, Macioce P, Visentin S, Ambrosini E. Human iPSC-Derived Astrocytes: A Powerful Tool to Study Primary Astrocyte Dysfunction in the Pathogenesis of Rare Leukodystrophies. Int J Mol Sci 2021; 23:ijms23010274. [PMID: 35008700 PMCID: PMC8745131 DOI: 10.3390/ijms23010274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are very versatile cells, endowed with multitasking capacities to ensure brain homeostasis maintenance from brain development to adult life. It has become increasingly evident that astrocytes play a central role in many central nervous system pathologies, not only as regulators of defensive responses against brain insults but also as primary culprits of the disease onset and progression. This is particularly evident in some rare leukodystrophies (LDs) where white matter/myelin deterioration is due to primary astrocyte dysfunctions. Understanding the molecular defects causing these LDs may help clarify astrocyte contribution to myelin formation/maintenance and favor the identification of possible therapeutic targets for LDs and other CNS demyelinating diseases. To date, the pathogenic mechanisms of these LDs are poorly known due to the rarity of the pathological tissue and the failure of the animal models to fully recapitulate the human diseases. Thus, the development of human induced pluripotent stem cells (hiPSC) from patient fibroblasts and their differentiation into astrocytes is a promising approach to overcome these issues. In this review, we discuss the primary role of astrocytes in LD pathogenesis, the experimental models currently available and the advantages, future evolutions, perspectives, and limitations of hiPSC to study pathologies implying astrocyte dysfunctions.
Collapse
Affiliation(s)
- Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Maria Stefania Brignone
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00169 Rome, Italy;
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
- Correspondence: ; Tel.: +39-064-990-2037
| |
Collapse
|
8
|
Light-stimulus intensity modulates startle reflex habituation in larval zebrafish. Sci Rep 2021; 11:22410. [PMID: 34789729 PMCID: PMC8599482 DOI: 10.1038/s41598-021-00535-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
The startle reflex in larval zebrafish describes a C-bend of the body occurring in response to sudden, unexpected, stimuli of different sensory modalities. Alterations in the startle reflex habituation (SRH) have been reported in various human and animal models of neurological and psychiatric conditions and are hence considered an important behavioural marker of neurophysiological function. The amplitude, offset and decay constant of the auditory SRH in larval zebrafish have recently been characterised, revealing that the measures are affected by variation in vibratory frequency, intensity, and interstimulus-interval. Currently, no study provides a model-based analysis of the effect of physical properties of light stimuli on the visual SRH. This study assessed the effect of incremental light-stimulus intensity on the SRH of larval zebrafish through a repeated-measures design. Their total locomotor responses were normalised for the time factor, based on the behaviour of a (non-stimulated) control group. A linear regression indicated that light intensity positively predicts locomotor responses due to larger SRH decay constants and offsets. The conclusions of this study provide important insights as to the effect of light properties on the SRH in larval zebrafish. Our methodology and findings constitute a relevant reference framework for further investigation in translational neurophysiological research.
Collapse
|
9
|
Alonso-Gardón M, Elorza-Vidal X, Castellanos A, La Sala G, Armand-Ugon M, Gilbert A, Di Pietro C, Pla-Casillanis A, Ciruela F, Gasull X, Nunes V, Martínez A, Schulte U, Cohen-Salmon M, Marazziti D, Estévez R. Identification of the GlialCAM interactome: the G protein-coupled receptors GPRC5B and GPR37L1 modulate megalencephalic leukoencephalopathy proteins. Hum Mol Genet 2021; 30:1649-1665. [PMID: 34100078 PMCID: PMC8369841 DOI: 10.1093/hmg/ddab155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/23/2022] Open
Abstract
Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events.
Collapse
Affiliation(s)
- Marta Alonso-Gardón
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Xabier Elorza-Vidal
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Aida Castellanos
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Mercedes Armand-Ugon
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris F-75005, France
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Adrià Pla-Casillanis
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, L'Hospitalet de Llobregat, Barcelona 08036, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, University of Barcelona-IDIBAPS, Casanova 143 Barcelona 08036, Spain
| | - Virginia Nunes
- Unitat de Genètica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Laboratori de Genètica Molecular, Genes Disease and Therapy Program IDIBELL, L'Hospitalet de Llobregat 08036, Spain
| | - Albert Martínez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | | | - Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris F-75005, France
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Raúl Estévez
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
10
|
Quint WH, Tadema KCD, de Vrieze E, Lukowicz RM, Broekman S, Winkelman BHJ, Hoevenaars M, de Gruiter HM, van Wijk E, Schaeffel F, Meester-Smoor M, Miller AC, Willemsen R, Klaver CCW, Iglesias AI. Loss of Gap Junction Delta-2 (GJD2) gene orthologs leads to refractive error in zebrafish. Commun Biol 2021; 4:676. [PMID: 34083742 PMCID: PMC8175550 DOI: 10.1038/s42003-021-02185-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Myopia is the most common developmental disorder of juvenile eyes, and it has become an increasing cause of severe visual impairment. The GJD2 locus has been consistently associated with myopia in multiple independent genome-wide association studies. However, despite the strong genetic evidence, little is known about the functional role of GJD2 in refractive error development. Here, we find that depletion of gjd2a (Cx35.5) or gjd2b (Cx35.1) orthologs in zebrafish, cause changes in the biometry and refractive status of the eye. Our immunohistological and scRNA sequencing studies show that Cx35.5 (gjd2a) is a retinal connexin and its depletion leads to hyperopia and electrophysiological changes in the retina. These findings support a role for Cx35.5 (gjd2a) in the regulation of ocular biometry. Cx35.1 (gjd2b) has previously been identified in the retina, however, we found an additional lenticular role. Lack of Cx35.1 (gjd2b) led to a nuclear cataract that triggered axial elongation. Our results provide functional evidence of a link between gjd2 and refractive error. Quint et al. use zebrafish lines deficient in one of two orthologs of the Gap Junction Delta-2 (GJD2) gene, which is associated with myopia by genome-wide association studies. They link gjd2 with refractive error and report evidence to suggest that gjd2a plays a role in ocular biometry whilst gjd2b, previously found in the retina, possesses an additional lenticular role.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands. .,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rachel M Lukowicz
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Sanne Broekman
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Beerend H J Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Melanie Hoevenaars
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Erwin van Wijk
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank Schaeffel
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Magda Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adam C Miller
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.,Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands. .,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Bosch A, Estévez R. Megalencephalic Leukoencephalopathy: Insights Into Pathophysiology and Perspectives for Therapy. Front Cell Neurosci 2021; 14:627887. [PMID: 33551753 PMCID: PMC7862579 DOI: 10.3389/fncel.2020.627887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/30/2020] [Indexed: 01/13/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare genetic disorder belonging to the group of vacuolating leukodystrophies. It is characterized by megalencephaly, loss of motor functions, epilepsy, and mild mental decline. In brain biopsies of MLC patients, vacuoles were observed in myelin and in astrocytes surrounding blood vessels. It is mainly caused by recessive mutations in MLC1 and HEPACAM (also called GLIALCAM) genes. These disease variants are called MLC1 and MLC2A with both types of patients sharing the same clinical phenotype. Besides, dominant mutations in HEPACAM were also identified in a subtype of MLC patients (MLC2B) with a remitting phenotype. MLC1 and GlialCAM proteins form a complex mainly expressed in brain astrocytes at the gliovascular interface and in Bergmann glia at the cerebellum. Both proteins regulate several ion channels and transporters involved in the control of ion and water fluxes in glial cells, either directly influencing their location and function, or indirectly regulating associated signal transduction pathways. However, the MLC1/GLIALCAM complex function and the related pathological mechanisms leading to MLC are still unknown. It has been hypothesized that, in MLC, the role of glial cells in brain ion homeostasis is altered in both physiological and inflammatory conditions. There is no therapy for MLC patients, only supportive treatment. As MLC2B patients show an MLC reversible phenotype, we speculated that the phenotype of MLC1 and MLC2A patients could also be mitigated by the re-introduction of the correct gene even at later stages. To prove this hypothesis, we injected in the cerebellar subarachnoid space of Mlc1 knockout mice an adeno-associated virus (AAV) coding for human MLC1 under the control of the glial-fibrillary acidic protein promoter. MLC1 expression in the cerebellum extremely reduced myelin vacuolation at all ages in a dose-dependent manner. This study could be considered as the first preclinical approach for MLC. We also suggest other potential therapeutic strategies in this review.
Collapse
Affiliation(s)
- Assumpció Bosch
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Univ. Autònoma de Barcelona, Barcelona, Spain.,Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Raúl Estévez
- Departament de Ciències Fisiològiques, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Sánchez A, García-Lareu B, Puig M, Prat E, Ruberte J, Chillón M, Nunes V, Estévez R, Bosch A. Cerebellar Astrocyte Transduction as Gene Therapy for Megalencephalic Leukoencephalopathy. Neurotherapeutics 2020; 17:2041-2053. [PMID: 32372403 PMCID: PMC7851290 DOI: 10.1007/s13311-020-00865-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare genetic disorder belonging to the group of vacuolating leukodystrophies. It is characterized by megalencephaly, loss of motor functions, epilepsy, and mild mental decline. In brain biopsies of MLC patients, vacuoles were observed in myelin and in astrocytes surrounding blood vessels. There is no therapy for MLC patients, only supportive treatment. We show here a preclinical gene therapy approach for MLC using the Mlc1 knock-out mouse. An adeno-associated virus coding for human MLC1 under the control of the glial fibrillary acidic protein promoter was injected in the cerebellar subarachnoid space of Mlc1 knock-out and wild-type animals at 2 months of age, before the onset of the disease, as a preventive approach. We also tested a therapeutic strategy by injecting the animals at 5 months, once the histopathological abnormalities are starting, or at 15 months, when they have progressed to a more severe pathology. MLC1 expression in the cerebellum restored the adhesion molecule GlialCAM and the chloride channel ClC-2 localization in Bergmann glia, which both are mislocalized in Mlc1 knock-out model. More importantly, myelin vacuolation was extremely reduced in treated mice at all ages and correlated with the amount of expressed MLC1 in Bergmann glia, indicating not only the preventive potential of this strategy but also its therapeutic capacity. In summary, here we provide the first therapeutic approach for patients affected with MLC. This work may have also implications to treat other diseases affecting motor function such as ataxias.
Collapse
Affiliation(s)
- Angela Sánchez
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Edifici H, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Belén García-Lareu
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Edifici H, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Spain
| | - Meritxell Puig
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Edifici H, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Esther Prat
- Laboratori de Genètica Molecular, Programa de Genes, Malaltia i Teràpia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Unitat de Genètica, Departament de Ciències Fisiològiques, Facultad de Medicina i Ciències de la Salut, Univ. de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Ruberte
- Department of Animal Health and Anatomy and Center of Animal Biotechnology and Gene Therapy (CBATEG), Univ. Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Chillón
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Edifici H, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Institut Català de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Virginia Nunes
- Laboratori de Genètica Molecular, Programa de Genes, Malaltia i Teràpia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Unitat de Genètica, Departament de Ciències Fisiològiques, Facultad de Medicina i Ciències de la Salut, Univ. de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Raul Estévez
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Departament de Ciències Fisiològiques, IDIBELL - Institute of Neurosciences, Universitat de Barcelona, E-08907, Barcelona, Spain.
| | - Assumpció Bosch
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Edifici H, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Spain.
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
13
|
Aladeokin AC, Akiyama T, Kimura A, Kimura Y, Takahashi-Jitsuki A, Nakamura H, Makihara H, Masukawa D, Nakabayashi J, Hirano H, Nakamura F, Saito T, Saido T, Goshima Y. Network-guided analysis of hippocampal proteome identifies novel proteins that colocalize with Aβ in a mice model of early-stage Alzheimer’s disease. Neurobiol Dis 2019; 132:104603. [DOI: 10.1016/j.nbd.2019.104603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/12/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022] Open
|
14
|
Pérez-Rius C, Folgueira M, Elorza-Vidal X, Alia A, Hoegg-Beiler MB, Eeza MNH, Díaz ML, Nunes V, Barrallo-Gimeno A, Estévez R. Comparison of zebrafish and mice knockouts for Megalencephalic Leukoencephalopathy proteins indicates that GlialCAM/MLC1 forms a functional unit. Orphanet J Rare Dis 2019; 14:268. [PMID: 31752924 PMCID: PMC6873532 DOI: 10.1186/s13023-019-1248-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/01/2019] [Indexed: 01/24/2023] Open
Abstract
Background Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a rare type of leukodystrophy characterized by astrocyte and myelin vacuolization, epilepsy and early-onset macrocephaly. MLC is caused by mutations in MLC1 or GLIALCAM, coding for two membrane proteins with an unknown function that form a complex specifically expressed in astrocytes at cell-cell junctions. Recent studies in Mlc1−/− or Glialcam−/− mice and mlc1−/− zebrafish have shown that MLC1 regulates glial surface levels of GlialCAM in vivo and that GlialCAM is also required for MLC1 expression and localization at cell-cell junctions. Methods We have generated and analysed glialcama−/− zebrafish. We also generated zebrafish glialcama−/−mlc1−/− and mice double KO for both genes and performed magnetic resonance imaging, histological studies and biochemical analyses. Results glialcama−/− shows megalencephaly and increased fluid accumulation. In both zebrafish and mice, this phenotype is not aggravated by additional elimination of mlc1. Unlike mice, mlc1 protein expression and localization are unaltered in glialcama−/− zebrafish, possibly because there is an up-regulation of mlc1 mRNA. In line with these results, MLC1 overexpressed in Glialcam−/− mouse primary astrocytes is located at cell-cell junctions. Conclusions This work indicates that the two proteins involved in the pathogenesis of MLC, GlialCAM and MLC1, form a functional unit, and thus, that loss-of-function mutations in these genes cause leukodystrophy through a common pathway.
Collapse
Affiliation(s)
- Carla Pérez-Rius
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mónica Folgueira
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008-A, Coruña, Spain.,Centro de Investigaciones Cientificas Avanzadas (CICA), University of A Coruña, 15008-A, Coruña, Spain
| | - Xabier Elorza-Vidal
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - A Alia
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.,Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Maja B Hoegg-Beiler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department Physiology and Pathology of Ion Transport, D-13125, Berlin, Germany.,Max-Delbruck-Centrum für Molekulare Medizin (MDC), D-13125, Berlin, Germany
| | - Muhamed N H Eeza
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - María Luz Díaz
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008-A, Coruña, Spain.,Centro de Investigaciones Cientificas Avanzadas (CICA), University of A Coruña, 15008-A, Coruña, Spain
| | - Virginia Nunes
- Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain.,Unitat de Genètica, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alejandro Barrallo-Gimeno
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| | - Raúl Estévez
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain. .,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain. .,Facultat de Medicina, Departament de Ciències Fisiològiques, Universitat de Barcelona-IDIBELL, C/Feixa Llarga s/n 08907 L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
15
|
Abstract
Leukodystrophies are genetically determined disorders affecting the white matter of the central nervous system. The combination of MRI pattern recognition and next-generation sequencing for the definition of novel disease entities has recently demonstrated that many leukodystrophies are due to the primary involvement and/or mutations in genes selectively expressed by cell types other than the oligodendrocytes, the myelin-forming cells in the brain. This has led to a new definition of leukodystrophies as genetic white matter disorders resulting from the involvement of any white matter structural component. As a result, the research has shifted its main focus from oligodendrocytes to other types of neuroglia. Astrocytes are the housekeeping cells of the nervous system, responsible for maintaining homeostasis and normal brain physiology and to orchestrate repair upon injury. Several lines of evidence show that astrocytic interactions with the other white matter cellular constituents play a primary pathophysiologic role in many leukodystrophies. These are thus now classified as astrocytopathies. This chapter addresses how the crosstalk between astrocytes, other glial cells, axons and non-neural cells are essential for the integrity and maintenance of the white matter in health. It also addresses the current knowledge of the cellular pathomechanisms of astrocytic leukodystrophies, and specifically Alexander disease, vanishing white matter, megalencephalic leukoencephalopathy with subcortical cysts and Aicardi-Goutière Syndrome.
Collapse
Affiliation(s)
- M S Jorge
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Girl With Progressive Head Enlargement and Gait Disturbance: Clinicoradiological Clues. Pediatr Neurol 2019; 97:80-81. [PMID: 30808533 DOI: 10.1016/j.pediatrneurol.2019.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 01/13/2019] [Accepted: 01/16/2019] [Indexed: 11/20/2022]
|
17
|
Gilbert A, Vidal XE, Estevez R, Cohen-Salmon M, Boulay AC. Postnatal development of the astrocyte perivascular MLC1/GlialCAM complex defines a temporal window for the gliovascular unit maturation. Brain Struct Funct 2019; 224:1267-1278. [PMID: 30684007 DOI: 10.1007/s00429-019-01832-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes, the most abundant glial cells of the central nervous system are morphologically complex. They display numerous processes interacting with synapses and blood vessels. At the vascular interface, astrocyte endfeet-terminated processes almost entirely cover the blood vessel surface and participate to the gliovascular unit where important vascular properties of the brain are set such as the blood-brain barrier (BBB) integrity. How specific morphological and functional interactions between astrocytes and the vascular compartment develop has not been fully investigated. Here, we elaborated an original experimental strategy to study the postnatal development of astrocyte perivascular endfeet. Using purified gliovascular units, we focused on the postnatal expression of MLC1 and GlialCAM, two transmembrane proteins forming a complex enriched at the junction between mature astrocyte perivascular endfeet. We showed that MLC1 and GlialCAM were enriched and assembled into mature complexes in astrocyte perivascular endfeet between postnatal days 10 and 15, after the formation of astrocyte perivascular Aquaporin 4 water channels. These events correlated with the increased expression of Claudin-5 and P-gP, two endothelial-specific BBB components. These results illustrate for the first time that astrocyte perivascular endfeet differentiation is a complex and progressive process which correlates with BBB maturation. Moreover, our results suggest that maturation of the astrocyte endfeet MLC1/GlialCAM complex between postnatal days 10 and 15 might be a key event in the gliovascular unit maturation.
Collapse
Affiliation(s)
- Alice Gilbert
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale INSERM, U1050, 11 place Marcelin Berthelot Paris, Paris Cedex 05, 75005, France
- Paris Science Lettre Research University, Paris, 75005, France
| | - Xabier Elorza Vidal
- Unitat de Fisiología, Departament de Ciències Fisiològiques, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Raul Estevez
- Unitat de Fisiología, Departament de Ciències Fisiològiques, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale INSERM, U1050, 11 place Marcelin Berthelot Paris, Paris Cedex 05, 75005, France.
- Paris Science Lettre Research University, Paris, 75005, France.
| | - Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale INSERM, U1050, 11 place Marcelin Berthelot Paris, Paris Cedex 05, 75005, France
- Paris Science Lettre Research University, Paris, 75005, France
| |
Collapse
|
18
|
Min R, van der Knaap MS. Genetic defects disrupting glial ion and water homeostasis in the brain. Brain Pathol 2019; 28:372-387. [PMID: 29740942 PMCID: PMC8028498 DOI: 10.1111/bpa.12602] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/02/2018] [Indexed: 12/23/2022] Open
Abstract
Electrical activity of neurons in the brain, caused by the movement of ions between intracellular and extracellular compartments, is the basis of all our thoughts and actions. Maintaining the correct ionic concentration gradients is therefore crucial for brain functioning. Ion fluxes are accompanied by the displacement of osmotically obliged water. Since even minor brain swelling leads to severe brain damage and even death, brain ion and water movement has to be tightly regulated. Glial cells, in particular astrocytes, play a key role in ion and water homeostasis. They are endowed with specific channels, pumps and carriers to regulate ion and water flow. Glial cells form a large panglial syncytium to aid the uptake and dispersal of ions and water, and make extensive contacts with brain fluid barriers for disposal of excess ions and water. Genetic defects in glial proteins involved in ion and water homeostasis disrupt brain functioning, thereby leading to neurological diseases. Since white matter edema is often a hallmark disease feature, many of these diseases are characterized as leukodystrophies. In this review we summarize our current understanding of inherited glial diseases characterized by disturbed brain ion and water homeostasis by integrating findings from MRI, genetics, neuropathology and animal models for disease. We discuss how mutations in different glial proteins lead to disease, and highlight the similarities and differences between these diseases. To come to effective therapies for this group of diseases, a better mechanistic understanding of how glial cells shape ion and water movement in the brain is crucial.
Collapse
Affiliation(s)
- Rogier Min
- Department of Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands.,Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Elorza-Vidal X, Sirisi S, Gaitán-Peñas H, Pérez-Rius C, Alonso-Gardón M, Armand-Ugón M, Lanciotti A, Brignone MS, Prat E, Nunes V, Ambrosini E, Gasull X, Estévez R. GlialCAM/MLC1 modulates LRRC8/VRAC currents in an indirect manner: Implications for megalencephalic leukoencephalopathy. Neurobiol Dis 2018; 119:88-99. [DOI: 10.1016/j.nbd.2018.07.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/25/2018] [Accepted: 07/28/2018] [Indexed: 01/09/2023] Open
|
20
|
Messchaert M, Dona M, Broekman S, Peters TA, Corral-Serrano JC, Slijkerman RWN, van Wijk E, Collin RWJ. Eyes shut homolog is important for the maintenance of photoreceptor morphology and visual function in zebrafish. PLoS One 2018; 13:e0200789. [PMID: 30052645 PMCID: PMC6063403 DOI: 10.1371/journal.pone.0200789] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/03/2018] [Indexed: 12/27/2022] Open
Abstract
Mutations in eyes shut homolog (EYS), a gene predominantly expressed in the photoreceptor cells of the retina, are among the most frequent causes of autosomal recessive (ar) retinitis pigmentosa (RP), a progressive retinal disorder. Due to the absence of EYS in several rodent species and its retina-specific expression, still little is known about the exact function of EYS and the pathogenic mechanism underlying EYS-associated RP. We characterized eys in zebrafish, by RT-PCR analysis on zebrafish eye-derived RNA, which led to the identification of a 8,715 nucleotide coding sequence that is divided over 46 exons. The transcript is predicted to encode a 2,905-aa protein that contains 39 EGF-like domains and five laminin A G-like domains, which overall shows 33% identity with human EYS. To study the function of EYS, we generated a stable eysrmc101/rmc101 mutant zebrafish model using CRISPR/Cas9 technology. The introduced lesion is predicted to result in premature termination of protein synthesis and lead to loss of Eys function. Immunohistochemistry on retinal sections revealed that Eys localizes at the region of the connecting cilium and that both rhodopsin and cone transducin are mislocalized in the absence of Eys. Electroretinogram recordings showed diminished b-wave amplitudes in eysrmc101/rmc101 zebrafish (5 dpf) compared to age- and strain-matched wild-type larvae. In addition, decreased locomotor activity in response to light stimuli was observed in eys mutant larvae. Altogether, our study shows that absence of Eys leads to a disorganized retinal architecture and causes visual dysfunction in zebrafish.
Collapse
Affiliation(s)
- Muriël Messchaert
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Margo Dona
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne Broekman
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Theo A. Peters
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julio C. Corral-Serrano
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ralph W. N. Slijkerman
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erwin van Wijk
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob W. J. Collin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
21
|
C2orf71a/pcare1 is important for photoreceptor outer segment morphogenesis and visual function in zebrafish. Sci Rep 2018; 8:9675. [PMID: 29946172 PMCID: PMC6018674 DOI: 10.1038/s41598-018-27928-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/17/2018] [Indexed: 02/03/2023] Open
Abstract
Mutations in C2orf71 are causative for autosomal recessive retinitis pigmentosa and occasionally cone-rod dystrophy. We have recently discovered that the protein encoded by this gene is important for modulation of the ciliary membrane through the recruitment of an actin assembly module, and have therefore renamed the gene to PCARE (photoreceptor cilium actin regulator). Here, we report on the identification of two copies of the c2orf71/pcare gene in zebrafish, pcare1 and pcare2. To study the role of the gene most similar to human PCARE, pcare1, we have generated a stable pcare1 mutant zebrafish model (designated pcare1rmc100/rmc100) in which the coding sequence was disrupted using CRISPR/Cas9 technology. Retinas of both embryonic (5 dpf) and adult (6 mpf) pcare1rmc100/rmc100 zebrafish display a clear disorganization of photoreceptor outer segments, resembling the phenotype observed in Pcare−/− mice. Optokinetic response and visual motor response measurements indicated visual impairment in pcare1rmc100/rmc100 zebrafish larvae at 5 dpf. In addition, electroretinogram measurements showed decreased b-wave amplitudes in pcare1rmc100/rmc100 zebrafish as compared to age- and strain-matched wild-type larvae, indicating a defect in the transretinal current. Altogether, our data show that lack of pcare1 causes a retinal phenotype in zebrafish and indicate that the function of the PCARE gene is conserved across species.
Collapse
|
22
|
Dona M, Slijkerman R, Lerner K, Broekman S, Wegner J, Howat T, Peters T, Hetterschijt L, Boon N, de Vrieze E, Sorusch N, Wolfrum U, Kremer H, Neuhauss S, Zang J, Kamermans M, Westerfield M, Phillips J, van Wijk E. Usherin defects lead to early-onset retinal dysfunction in zebrafish. Exp Eye Res 2018; 173:148-159. [PMID: 29777677 DOI: 10.1016/j.exer.2018.05.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 01/24/2023]
Abstract
Mutations in USH2A are the most frequent cause of Usher syndrome and autosomal recessive nonsyndromic retinitis pigmentosa. To unravel the pathogenic mechanisms underlying USH2A-associated retinal degeneration and to evaluate future therapeutic strategies that could potentially halt the progression of this devastating disorder, an animal model is needed. The available Ush2a knock-out mouse model does not mimic the human phenotype, because it presents with only a mild and late-onset retinal degeneration. Using CRISPR/Cas9-technology, we introduced protein-truncating germline lesions into the zebrafish ush2a gene (ush2armc1: c.2337_2342delinsAC; p.Cys780GlnfsTer32 and ush2ab1245: c.15520_15523delinsTG; p.Ala5174fsTer). Homozygous mutants were viable and displayed no obvious morphological or developmental defects. Immunohistochemical analyses with antibodies recognizing the N- or C-terminal region of the ush2a-encoded protein, usherin, demonstrated complete absence of usherin in photoreceptors of ush2armc1, but presence of the ectodomain of usherin at the periciliary membrane of ush2ab1245-derived photoreceptors. Furthermore, defects of usherin led to a reduction in localization of USH2 complex members, whirlin and Adgrv1, at the photoreceptor periciliary membrane of both mutants. Significantly elevated levels of apoptotic photoreceptors could be observed in both mutants when kept under constant bright illumination for three days. Electroretinogram (ERG) recordings revealed a significant and similar decrease in both a- and b-wave amplitudes in ush2armc1 as well as ush2ab1245 larvae as compared to strain- and age-matched wild-type larvae. In conclusion, this study shows that mutant ush2a zebrafish models present with early-onset retinal dysfunction that is exacerbated by light exposure. These models provide a better understanding of the pathophysiology underlying USH2A-associated RP and a unique opportunity to evaluate future therapeutic strategies.
Collapse
Affiliation(s)
- Margo Dona
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Ralph Slijkerman
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Kimberly Lerner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Sanne Broekman
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Jeremy Wegner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Taylor Howat
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Theo Peters
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Lisette Hetterschijt
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Nanda Boon
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Nasrin Sorusch
- Institute of Molecular Physiology, Johannes Gutenberg University, Johannes-von-Muellerweg 6, D-55099 Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, Johannes-von-Muellerweg 6, D-55099 Mainz, Germany
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands
| | - Stephan Neuhauss
- University of Zürich, Institute of Molecular Life Sciences, Winterthurerstrasse 190, Zürich, CH - 8057, Switzerland
| | - Jingjing Zang
- University of Zürich, Institute of Molecular Life Sciences, Winterthurerstrasse 190, Zürich, CH - 8057, Switzerland
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; Department of Biomedical Physics, Academisch Medisch Centrum, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Monte Westerfield
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Jennifer Phillips
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403-1254, USA
| | - Erwin van Wijk
- Department of Otorhinolaryngology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behavior, Montessorilaan 3, 6525 HR Nijmegen, The Netherlands.
| |
Collapse
|
23
|
Estévez R, Elorza-Vidal X, Gaitán-Peñas H, Pérez-Rius C, Armand-Ugón M, Alonso-Gardón M, Xicoy-Espaulella E, Sirisi S, Arnedo T, Capdevila-Nortes X, López-Hernández T, Montolio M, Duarri A, Teijido O, Barrallo-Gimeno A, Palacín M, Nunes V. Megalencephalic leukoencephalopathy with subcortical cysts: A personal biochemical retrospective. Eur J Med Genet 2018; 61:50-60. [DOI: 10.1016/j.ejmg.2017.10.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/14/2017] [Accepted: 10/22/2017] [Indexed: 12/22/2022]
|
24
|
van der Knaap MS, Bugiani M. Leukodystrophies: a proposed classification system based on pathological changes and pathogenetic mechanisms. Acta Neuropathol 2017; 134:351-382. [PMID: 28638987 PMCID: PMC5563342 DOI: 10.1007/s00401-017-1739-1] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022]
Abstract
Leukodystrophies are genetically determined disorders characterized by the selective involvement of the central nervous system white matter. Onset may be at any age, from prenatal life to senescence. Many leukodystrophies are degenerative in nature, but some only impair white matter function. The clinical course is mostly progressive, but may also be static or even improving with time. Progressive leukodystrophies are often fatal, and no curative treatment is known. The last decade has witnessed a tremendous increase in the number of defined leukodystrophies also owing to a diagnostic approach combining magnetic resonance imaging pattern recognition and next generation sequencing. Knowledge on white matter physiology and pathology has also dramatically built up. This led to the recognition that only few leukodystrophies are due to mutations in myelin- or oligodendrocyte-specific genes, and many are rather caused by defects in other white matter structural components, including astrocytes, microglia, axons and blood vessels. We here propose a novel classification of leukodystrophies that takes into account the primary involvement of any white matter component. Categories in this classification are the myelin disorders due to a primary defect in oligodendrocytes or myelin (hypomyelinating and demyelinating leukodystrophies, leukodystrophies with myelin vacuolization); astrocytopathies; leuko-axonopathies; microgliopathies; and leuko-vasculopathies. Following this classification, we illustrate the neuropathology and disease mechanisms of some leukodystrophies taken as example for each category. Some leukodystrophies fall into more than one category. Given the complex molecular and cellular interplay underlying white matter pathology, recognition of the cellular pathology behind a disease becomes crucial in addressing possible treatment strategies.
Collapse
Affiliation(s)
- Marjo S van der Knaap
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Functional Genomics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Dai CL, He WB, Du J, Tan YQ, Lu GX, Li W. A case of megalencephalic leukoencephalopathy with subcortical cysts type 1 was identified with a novel compound heterozygous alteration (c.135delC; c.423+2dupT) in China. Clin Case Rep 2017; 5:961-967. [PMID: 28588848 PMCID: PMC5458048 DOI: 10.1002/ccr3.986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 11/08/2022] Open
Abstract
We report a compound heterozygous mutation (c.135delC; c.423+2dupT) of MLC1 gene in a Chinese patient underlying infantile macrocephaly and neurological deterioration in early childhood. Brain MRI revealed diffusion abnormality in swollen white matter and a subcortical cyst. The cDNA sequencing analysis for the c.423+2dupT variant revealed skipping of exon 5.
Collapse
Affiliation(s)
- Cong-Ling Dai
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China
| | - Wen-Bin He
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China.,Reproductive and Genetic Hospital of Citic-Xiangya Hunan 410008 China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China.,Reproductive and Genetic Hospital of Citic-Xiangya Hunan 410008 China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China.,Reproductive and Genetic Hospital of Citic-Xiangya Hunan 410008 China
| | - Guang-Xiu Lu
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China.,Reproductive and Genetic Hospital of Citic-Xiangya Hunan 410008 China.,National Engineering and Research Center of Human Stem Cells Changsha Hunan 410000 China
| | - Wen Li
- Institute of Reproductive and Stem Cell Engineering Central South University Hunan 410008 China.,Reproductive and Genetic Hospital of Citic-Xiangya Hunan 410008 China
| |
Collapse
|
26
|
Bugiani M, Dubey M, Breur M, Postma NL, Dekker MP, Ter Braak T, Boschert U, Abbink TEM, Mansvelder HD, Min R, van Weering JRT, van der Knaap MS. Megalencephalic leukoencephalopathy with cysts: the Glialcam-null mouse model. Ann Clin Transl Neurol 2017; 4:450-465. [PMID: 28695146 PMCID: PMC5497535 DOI: 10.1002/acn3.405] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/23/2022] Open
Abstract
Objective Megalencephalic leukoencephalopathy with cysts (MLC) is a genetic infantile‐onset disease characterized by macrocephaly and white matter edema due to loss of MLC1 function. Recessive mutations in either MLC1 or GLIALCAM cause the disease. MLC1 is involved in astrocytic volume regulation; GlialCAM ensures the correct membrane localization of MLC1. Their exact role in brain ion‐water homeostasis is only partly defined. We characterized Glialcam‐null mice for further studies. Methods We investigated the consequences of loss of GlialCAM in Glialcam‐null mice and compared GlialCAM developmental expression in mice and men. Results Glialcam‐null mice had early‐onset megalencephaly and increased brain water content. From 3 weeks, astrocytes were abnormal with swollen processes abutting blood vessels. Concomitantly, progressive white matter vacuolization developed due to intramyelinic edema. Glialcam‐null astrocytes showed abolished expression of MLC1, reduced expression of the chloride channel ClC‐2 and increased expression and redistribution of the water channel aquaporin4. Expression of other MLC1‐interacting proteins and the volume regulated anion channel LRRC8A was unchanged. In mice, GlialCAM expression increased until 3 weeks and then stabilized. In humans, GlialCAM expression was highest in the first 3 years to then decrease and stabilize from approximately 5 years. Interpretation Glialcam‐null mice replicate the early stages of the human disease with early‐onset intramyelinic edema. The earliest change is astrocytic swelling, further substantiating that a defect in astrocytic volume regulation is the primary cellular defect in MLC. GlialCAM expression affects expression of MLC1, ClC‐2 and aquaporin4, indicating that abnormal interplay between these proteins is a disease mechanism in megalencephalic leukoencephalopathy with cysts.
Collapse
Affiliation(s)
- Marianna Bugiani
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands.,Department of Pathology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands
| | - Mohit Dubey
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands.,Department of Integrative Neurophysiology Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| | - Marjolein Breur
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands
| | - Nienke L Postma
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands
| | - Marien P Dekker
- Department of Functional Genomics Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| | - Timo Ter Braak
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands
| | - Ursula Boschert
- Translational Innovation Platform Immunology/Neurology EMD Serono Research & Development Institute Billerica 01821 Massachusetts
| | - Truus E M Abbink
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| | - Rogier Min
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands.,Department of Integrative Neurophysiology Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| | - Jan R T van Weering
- Department of Functional Genomics Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| | - Marjo S van der Knaap
- Department of Pediatrics/Child Neurology Amsterdam Neuroscience VU University Medical Center Amsterdam The Netherlands.,Department of Functional Genomics Center for Neurogenomics and Cognitive Research Amsterdam Neuroscience VU University Amsterdam The Netherlands
| |
Collapse
|
27
|
Sirisi S, Elorza-Vidal X, Arnedo T, Armand-Ugón M, Callejo G, Capdevila-Nortes X, López-Hernández T, Schulte U, Barrallo-Gimeno A, Nunes V, Gasull X, Estévez R. Depolarization causes the formation of a ternary complex between GlialCAM, MLC1 and ClC-2 in astrocytes: implications in megalencephalic leukoencephalopathy. Hum Mol Genet 2017; 26:2436-2450. [DOI: 10.1093/hmg/ddx134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 01/06/2023] Open
|
28
|
Barrallo-Gimeno A, Gradogna A, Zanardi I, Pusch M, Estévez R. Regulatory-auxiliary subunits of CLC chloride channel-transport proteins. J Physiol 2016; 593:4111-27. [PMID: 25762128 DOI: 10.1113/jp270057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/15/2015] [Indexed: 02/06/2023] Open
Abstract
The CLC family of chloride channels and transporters is composed by nine members, but only three of them, ClC-Ka/b, ClC-7 and ClC-2, have been found so far associated with auxiliary subunits. These CLC regulatory subunits are small proteins that present few common characteristics among them, both structurally and functionally, and their effects on the corresponding CLC protein are different. Barttin, a protein with two transmembrane domains, is essential for the membrane localization of ClC-K proteins and their activity in the kidney and inner ear. Ostm1 is a protein with a single transmembrane domain and a highly glycosylated N-terminus. Unlike the other two CLC auxiliary subunits, Ostm1 shows a reciprocal relationship with ClC-7 for their stability. The subcellular localization of Ostm1 depends on ClC-7 and not the other way around. ClC-2 is active on its own, but GlialCAM, a transmembrane cell adhesion molecule with two extracellular immunoglobulin (Ig)-like domains, regulates its subcellular localization and activity in glial cells. The common theme for these three proteins is their requirement for a proper homeostasis, since their malfunction leads to distinct diseases. We will review here their properties and their role in normal chloride physiology and the pathological consequences of their improper function.
Collapse
Affiliation(s)
- Alejandro Barrallo-Gimeno
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, University of Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| | | | - Ilaria Zanardi
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genoa, Italy
| | - Michael Pusch
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genoa, Italy
| | - Raúl Estévez
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, University of Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| |
Collapse
|
29
|
Zang J, Keim J, Kastenhuber E, Gesemann M, Neuhauss SCF. Recoverin depletion accelerates cone photoresponse recovery. Open Biol 2016; 5:rsob.150086. [PMID: 26246494 PMCID: PMC4554923 DOI: 10.1098/rsob.150086] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The neuronal Ca2+-binding protein Recoverin has been shown to regulate phototransduction termination in mammalian rods. Here we identify four recoverin genes in the zebrafish genome, rcv1a, rcv1b, rcv2a and rcv2b, and investigate their role in modulating the cone phototransduction cascade. While Recoverin-1b is only found in the adult retina, the other Recoverins are expressed throughout development in all four cone types, except Recoverin-1a, which is expressed only in rods and UV cones. Applying a double flash electroretinogram (ERG) paradigm, downregulation of Recoverin-2a or 2b accelerates cone photoresponse recovery, albeit at different light intensities. Exclusive recording from UV cones via spectral ERG reveals that knockdown of Recoverin-1a alone has no effect, but Recoverin-1a/2a double-knockdowns showed an even shorter recovery time than Recoverin-2a-deficient larvae. We also showed that UV cone photoresponse kinetics depend on Recoverin-2a function via cone-specific kinase Grk7a. This is the first in vivo study demonstrating that cone opsin deactivation kinetics determine overall photoresponse shut off kinetics.
Collapse
Affiliation(s)
- Jingjing Zang
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Jennifer Keim
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Edda Kastenhuber
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Matthias Gesemann
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Stephan C F Neuhauss
- Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| |
Collapse
|
30
|
|
31
|
Zebrafish Models for the Mechanosensory Hair Cell Dysfunction in Usher Syndrome 3 Reveal That Clarin-1 Is an Essential Hair Bundle Protein. J Neurosci 2015; 35:10188-201. [PMID: 26180195 DOI: 10.1523/jneurosci.1096-15.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Usher syndrome type III (USH3) is characterized by progressive loss of hearing and vision, and varying degrees of vestibular dysfunction. It is caused by mutations that affect the human clarin-1 protein (hCLRN1), a member of the tetraspanin protein family. The missense mutation CLRN1(N48K), which affects a conserved N-glycosylation site in hCLRN1, is a common causative USH3 mutation among Ashkenazi Jews. The affected individuals hear at birth but lose that function over time. Here, we developed an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. Immunolabeling demonstrated that Clrn1 localized to the hair cell bundles (hair bundles). The clrn1 mutants generated by zinc finger nucleases displayed aberrant hair bundle morphology with diminished function. Two transgenic zebrafish that express either hCLRN1 or hCLRN1(N48K) in hair cells were produced to examine the subcellular localization patterns of wild-type and mutant human proteins. hCLRN1 localized to the hair bundles similarly to zebrafish Clrn1; in contrast, hCLRN1(N48K) largely mislocalized to the cell body with a small amount reaching the hair bundle. We propose that this small amount of hCLRN1(N48K) in the hair bundle provides clarin-1-mediated function during the early stages of life; however, the presence of hCLRN1(N48K) in the hair bundle diminishes over time because of intracellular degradation of the mutant protein, leading to progressive loss of hair bundle integrity and hair cell function. These findings and genetic tools provide an understanding and path forward to identify therapies to mitigate hearing loss linked to the CLRN1 mutation. SIGNIFICANCE STATEMENT Mutations in the clarin-1 gene affect eye and ear function in humans. Individuals with the CLRN1(N48K) mutation are born able to hear but lose that function over time. Here, we develop an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. This approach illuminates the role of clarin-1 and the molecular mechanism linked to the CLRN1(N48K) mutation in sensory hair cells of the inner ear. Additionally, the investigation provided an in vivo model to guide future drug discovery to rescue the hCLRN1(N48K) in hair cells.
Collapse
|
32
|
Capdevila-Nortes X, Jeworutzki E, Elorza-Vidal X, Barrallo-Gimeno A, Pusch M, Estévez R. Structural determinants of interaction, trafficking and function in the ClC-2/MLC1 subunit GlialCAM involved in leukodystrophy. J Physiol 2015; 593:4165-80. [PMID: 26033718 DOI: 10.1113/jp270467] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/22/2015] [Indexed: 02/03/2023] Open
Abstract
KEY POINTS The extracellular domain of GlialCAM is necessary for its targeting to cell junctions, as well as for interactions with itself and MLC1 and ClC-2. The C-terminus of GlialCAM is not necessary for interaction but is required for targeting to cell junctions. The first three residues of the transmembrane segment of GlialCAM are required for GlialCAM-mediated ClC-2 activation. ABSTRACT Mutations in the genes encoding the astrocytic protein MLC1, the cell adhesion molecule GlialCAM or the Cl(-) channel ClC-2 underlie human leukodystrophies. GlialCAM binds to itself, to MLC1 and to ClC-2, and directs these proteins to cell-cell contacts. In addition, GlialCAM dramatically activates ClC-2 mediated currents. In the present study, we used mutagenesis studies combined with functional and biochemical analyses to determine which parts of GlialCAM are required to perform these cellular functions. We found that the extracellular domain of GlialCAM is necessary for cell junction targeting and for mediating interactions with itself or with MLC1 and ClC-2. The C-terminus is also necessary for proper targeting to cell-cell junctions but is not required for the biochemical interaction. Finally, we identified the first three amino acids of the transmembrane segment of GlialCAM as being essential for the activation of ClC-2 currents but not for targeting or biochemical interaction. Our results provide new mechanistic insights concerning the regulation of the cell biology and function of MLC1 and ClC-2 by GlialCAM.
Collapse
Affiliation(s)
- Xavier Capdevila-Nortes
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, Universidad de Barcelona, Barcelona, Spain
| | - Elena Jeworutzki
- Istituto di Biofisica, CNR, Genoa, Italy.,Present address IfGH-Myocellular Electrophysiology, Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany
| | - Xabier Elorza-Vidal
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, Universidad de Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| | - Alejandro Barrallo-Gimeno
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, Universidad de Barcelona, Barcelona, Spain
| | | | - Raúl Estévez
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, Universidad de Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| |
Collapse
|
33
|
Brignone MS, Lanciotti A, Camerini S, De Nuccio C, Petrucci TC, Visentin S, Ambrosini E. MLC1 protein: a likely link between leukodystrophies and brain channelopathies. Front Cell Neurosci 2015; 9:66. [PMID: 25883547 PMCID: PMC4381631 DOI: 10.3389/fncel.2015.00106] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/09/2015] [Indexed: 01/12/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLCs) disease is a rare inherited, autosomal recessive form of childhood-onset spongiform leukodystrophy characterized by macrocephaly, deterioration of motor functions, epileptic seizures and mental decline. Brain edema, subcortical fluid cysts, myelin and astrocyte vacuolation are the histopathological hallmarks of MLC. Mutations in either the MLC1 gene (>75% of patients) or the GlialCAM gene (<20% of patients) are responsible for the disease. Recently, the GlialCAM adhesion protein was found essential for the membrane expression and function of the chloride channel ClC-2 indicating MLC disease caused by mutation in GlialCAM as the first channelopathy among leukodystrophies. On the contrary, the function of MLC1 protein, which binds GlialCAM, its functional relationship with ClC-2 and the molecular mechanisms underlying MLC1 mutation-induced functional defects are not fully understood yet. The human MLC1 gene encodes a 377-amino acid membrane protein with eight predicted transmembrane domains which shows very low homology with voltage-dependent potassium (K+) channel subunits. The high expression of MLC1 in brain astrocytes contacting blood vessels and meninges and brain alterations observed in MLC patients have led to hypothesize a role for MLC1 in the regulation of ion and water homeostasis. Recent studies have shown that MLC1 establishes structural and/or functional interactions with several ion/water channels and transporters and ion channel accessory proteins, and that these interactions are affected by MLC1 mutations causing MLC. Here, we review data on MLC1 functional properties obtained in in vitro and in vivo models and discuss evidence linking the effects of MLC1 mutations to brain channelopathies.
Collapse
Affiliation(s)
- Maria S Brignone
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Angela Lanciotti
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Serena Camerini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Chiara De Nuccio
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Tamara C Petrucci
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Sergio Visentin
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| | - Elena Ambrosini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Rome, Italy
| |
Collapse
|