1
|
Brao A, Sánchez Á, Rodríguez I, Rey JD, Lope-Piedrafita S, Prat E, Nunes V, Chillón M, Estévez R, Bosch A. Gene therapy rescues brain edema and motor function in a mouse model of megalencephalic leukoencephalopathy with subcortical cysts. Mol Ther 2025; 33:1434-1448. [PMID: 40051162 PMCID: PMC11997501 DOI: 10.1016/j.ymthe.2025.02.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is an ultrarare, infantile-onset leukodystrophy characterized by white matter edema for which there is no treatment. More than 75% of diagnosed cases result from biallelic loss-of-function mutations in the astrocyte-specific gene MLC1, leading to early-onset macrocephaly, cerebellar ataxia, epilepsy, and mild cognitive decline. To develop a gene therapy for MLC, we administered an adeno-associated viral vector capable of crossing the murine blood-brain barrier, delivering the human MLC1 cDNA under the control of a human astrocyte-specific promoter, to 10-month-old Mlc1-/- mice. We observed long-term astrocyte-driven expression of MLC1 up to 1 year after viral vector administration in all brain areas analyzed. Despite the late-stage intervention, in vivo magnetic resonance imaging revealed normalization of water accumulation. Notably, our therapy successfully reversed locomotor deficits in Mlc1-/- mice, as evidenced by improved performance in motor tests assessing cerebellar ataxia-like behaviors. Collectively, these findings not only demonstrate the sustained efficacy of our gene therapy but also highlight the reversibility of vacuolation and motor impairments in Mlc1-/- mice, suggesting that MLC patients could benefit from treatment even after symptom onset.
Collapse
Affiliation(s)
- Alejandro Brao
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Ángela Sánchez
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Irina Rodríguez
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Javier Del Rey
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Silvia Lope-Piedrafita
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Nuclear Magnetic Resonance Service, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Esther Prat
- Department of Physiological Sciences, Institute of Neurosciences, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, 08908 L'Hospitalet de Llobregat, Spain
| | - Virginia Nunes
- Department of Physiological Sciences, Institute of Neurosciences, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, 08908 L'Hospitalet de Llobregat, Spain
| | - Miguel Chillón
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Raúl Estévez
- Department of Physiological Sciences, Institute of Neurosciences, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, 08908 L'Hospitalet de Llobregat, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Assumpció Bosch
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; UAB-VHIR Joint Unit, Vall d'Hebron Institut de Recerca, 08035 Barcelona, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, 28031 Madrid, Spain.
| |
Collapse
|
2
|
Sabatino JJ, Steinman L. Multiple Sclerosis: A Virus-Induced Gliopathy? NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200383. [PMID: 39933124 DOI: 10.1212/nxi.0000000000200383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 02/13/2025]
Affiliation(s)
- Joseph J Sabatino
- Department of Neurology, University of California, San Francisco
- UCSF Weill Institute for Neurosciences; and
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA
| |
Collapse
|
3
|
Dahl JR, Weier A, Winter C, Hintze M, Rothhammer V, Tsaktanis T, Proebstel AK, Neziraj T, Poessnecker E, Oechtering J, Kuhle J, Kallmann BA, Luber G, Heider T, Klotz L, Chunder R, Kuerten S. Modulator of VRAC Current 1 Is a Potential Target Antigen in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200374. [PMID: 39933126 PMCID: PMC11839221 DOI: 10.1212/nxi.0000000000200374] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND AND OBJECTIVES Multiple sclerosis (MS) is a chronic immune-mediated demyelinating disease of the CNS. Highlighted by the success of B-cell-depleting therapies such as the monoclonal anti-CD20 antibodies rituximab, ocrelizumab, and ofatumumab, B cells have been shown to play a central role in the immunopathology of the disease. Yet, the target antigens of the pathogenic B-cell response in MS remain unclear. METHODS We combined polyclonal B-cell stimulation of peripheral blood mononuclear cells with a human proteome-wide protein microarray to identify target antigens of MS by comparing samples from 20 patients with MS with 9 age-matched and sex-matched healthy controls. Results were verified by enzyme-linked immunosorbent assay (ELISA) in 3 independent validation cohorts (N = 47 patients with MS in remission; N = 20 patients with MS during relapse; N = 25 HCs; N = 30 patients with other noninflammatory neurologic diseases; N = 9 patients with other inflammatory neurologic diseases). Experimental autoimmune encephalomyelitis (EAE) was used as an animal model to evaluate the pathogenicity of the antibodies of choice. RESULTS Our results corroborate the existing concept of a highly diverse autoimmune response in MS. Yet, a significantly elevated antibody response against the membrane protein modulator of VRAC current 1 (MLC1) was noted in B-cell culture supernatants and serum samples of patients with MS. Furthermore, significantly elevated titers to MLC1 were observed in the CSF of patients with neuroinflammatory diseases other than MS. Neurons and astrocytes were identified as the main cell types expressing MLC1 in the brain of a patient with MS. Injection of anti-MLC1 antibodies into mice with EAE led to strong in vivo binding to cerebral cortical neurons and to the death of 4 of the 7 injected mice. DISCUSSION Future studies will have to address the diagnostic and prognostic value of MLC1-specific antibodies in neuroinflammatory disorders such as MS and characterize the functional role of MLC1 expression in neurons and astrocytes. TRIAL REGISTRATION INFORMATION The study has been registered in the German Clinical Trials Register (study number DRKS00015528).
Collapse
Affiliation(s)
- Johannes Raffael Dahl
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn and University Hospital Bonn, Germany
| | - Alicia Weier
- Institute of Neuroanatomy, Medical Faculty, University of Bonn and University Hospital Bonn, Germany
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Australia
| | | | - Maik Hintze
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn and University Hospital Bonn, Germany
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Germany
| | | | - Anne-Katrin Proebstel
- Department of Neurology, University Hospital of Basel and University of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Switzerland
| | - Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Switzerland
| | - Elisabeth Poessnecker
- Department of Neurology, University Hospital of Basel and University of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Switzerland
| | - Johanna Oechtering
- Department of Neurology, University Hospital of Basel and University of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Switzerland
| | - Jens Kuhle
- Department of Neurology, University Hospital of Basel and University of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Switzerland
| | | | - Gabriele Luber
- Practice for Neurology, Psychiatry and Psychotherapy, Nürnberg, Germany
| | - Thorsten Heider
- Department of Neurology, Klinikum St. Marien Amberg, Germany; and
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University of Münster, Germany
| | - Rittika Chunder
- Institute of Neuroanatomy, Medical Faculty, University of Bonn and University Hospital Bonn, Germany
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Medical Faculty, University of Bonn and University Hospital Bonn, Germany
| |
Collapse
|
4
|
Sharma S, Bharti V, Das PK, Rahman A, Sharma H, Rauthan R, Rc M, Gupta N, Shukla R, Mohanty S, Kabra M, Francis KR, Chakraborty D. MLC1 alteration in iPSCs give rise to disease-like cellular vacuolation phenotype in the astrocyte lineage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631607. [PMID: 39829899 PMCID: PMC11741324 DOI: 10.1101/2025.01.06.631607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare and progressive neurodegenerative disorder involving the white matter, is not adequately recapitulated by current disease models. Somatic cell reprogramming, along with advancements in genome engineering, may allow the establishment of in-vitro human models of MLC for disease modeling and drug screening. In this study, we utilized cellular reprogramming and gene-editing techniques to develop induced pluripotent stem cell (iPSC) models of MLC to recapitulate the cellular context of the classical MLC-impacted nervous system. Methods Somatic cell reprogramming of peripheral patient-derived blood mononuclear cells (PBMCs) was used to develop iPSC models of MLC. CRISPR-Cas9 system-based genome engineering was also utilized to create the MLC1 knockout model of the disease. Directed differentiation of iPSCs to neural stem cells (NSCs) and astrocytes was performed in a 2D cell culture format, followed by various cellular and molecular biology approaches, to characterize the disease model. Results MLC iPSCs established by somatic cell reprogramming and genome engineering were well characterized for pluripotency. iPSCs were subsequently differentiated to disease-relevant cell types: neural stem cells (NSCs) and astrocytes. RNA sequencing profiling of MLC NSCs revealed a set of differentially expressed genes related to neurological disorders and epilepsy, a common clinical finding within MLC disease. This gene set can serve as a target for drug screening for the development of a potential therapeutic for this disease. Upon differentiation to the more disease relevant cell type-astrocytes, MLC-characteristic vacuoles were clearly observed, which were distinctly absent from controls. This emergence recapitulated a distinguishing phenotypic marker of the disease. Conclusion Through the creation and analyses of iPSC models of MLC, our work addresses a critical need for relevant cellular models of MLC for use in both disease modeling and drug screening assays. Further investigation can utilize MLC iPSC models, as well as generated transcriptomic data sets and analyses, to identify potential therapeutic interventions for this debilitating disease.
Collapse
|
5
|
Hol EM, Dykstra W, Chevalier J, Cuadrado E, Bugiani M, Aronica E, Verkhratsky A. Neuroglia in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:159-175. [PMID: 40148043 DOI: 10.1016/b978-0-443-19102-2.00032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Leukodystrophies are a heterogeneous group of rare genetic neurologic disorders characterized by white matter degeneration resulting from mutations affecting glial cells. This review focuses on the primary subtypes-astroglial, oligodendroglial, and microglial leukodystrophies-offering a detailed description of their neuropathologic features and clinical manifestations. It delves into key aspects of the pathogenesis, emphasizing the distinct cellular mechanisms that drive white matter damage. Advances in disease modeling, including the development of animal models with pathologic gene expressions and patient-derived iPS-cell models, have significantly enhanced our understanding of these rare disorders. Insights into the roles of different glial cell types highlight the complexity of leukodystrophies and provide a foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Juliette Chevalier
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eloy Cuadrado
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
6
|
Schrader JM, Xu F, Agostinucci KJ, DaSilva NA, Van Nostrand WE. Longitudinal markers of cerebral amyloid angiopathy and related inflammation in rTg-DI rats. Sci Rep 2024; 14:8441. [PMID: 38600214 PMCID: PMC11006668 DOI: 10.1038/s41598-024-59013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 04/05/2024] [Indexed: 04/12/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a prevalent vascular dementia and common comorbidity of Alzheimer's disease (AD). While it is known that vascular fibrillar amyloid β (Aβ) deposits leads to vascular deterioration and can drive parenchymal CAA related inflammation (CAA-ri), underlying mechanisms of CAA pathology remain poorly understood. Here, we conducted brain regional proteomic analysis of early and late disease stages in the rTg-DI CAA rat model to gain molecular insight to mechanisms of CAA/CAA-ri progression and identify potential brain protein markers of CAA/CAA-ri. Longitudinal brain regional proteomic analysis revealed increased differentially expressed proteins (DEP) including ANXA3, HTRA1, APOE, CST3, and CLU, shared between the cortex, hippocampus, and thalamus, at both stages of disease in rTg-DI rats. Subsequent pathway analysis indicated pathway enrichment and predicted activation of TGF-β1, which was confirmed by immunolabeling and ELISA. Further, we identified numerous CAA related DEPs associate with astrocytes (HSPB1 and MLC1) and microglia (ANXA3, SPARC, TGF-β1) not previously associated with astrocytes or microglia in other AD models, possibly indicating that they are specific to CAA-ri. Thus, the data presented here identify several potential brain protein biomarkers of CAA/CAA-ri while providing novel molecular and mechanistic insight to mechanisms of CAA and CAA-ri pathological progression and glial cell mediated responses.
Collapse
Affiliation(s)
- Joseph M Schrader
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, Rhode Island, 02881, USA
| | - Feng Xu
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, Rhode Island, 02881, USA
| | - Kevin J Agostinucci
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, Rhode Island, 02881, USA
| | - Nicholas A DaSilva
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, 02912, USA
| | - William E Van Nostrand
- Department of Biomedical and Pharmaceutical Sciences, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, Rhode Island, 02881, USA.
| |
Collapse
|
7
|
Kaur N, Arora K, Radhakrishnan P, Narayanan DL, Shukla A. Intragenic homozygous duplication in HEPACAM is associated with megalencephalic leukoencephalopathy with subcortical cysts type 2A. Neurogenetics 2024; 25:85-91. [PMID: 38280046 DOI: 10.1007/s10048-024-00743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/05/2024] [Indexed: 01/29/2024]
Abstract
Disease-causing variants in HEPACAM are associated with megalencephalic leukoencephalopathy with subcortical cysts 2A (MLC2A, MIM# 613,925, autosomal recessive), and megalencephalic leukoencephalopathy with subcortical cysts 2B, remitting, with or without impaired intellectual development (MLC2B, MIM# 613,926, autosomal dominant). These disorders are characterised by macrocephaly, seizures, motor delay, cognitive impairment, ataxia, and spasticity. Brain magnetic resonance imaging (MRI) in these individuals shows swollen cerebral hemispheric white matter and subcortical cysts, mainly in the frontal and temporal regions. To date, 45 individuals from 39 families are reported with biallelic and heterozygous variants in HEPACAM, causing MLC2A and MLC2B, respectively. A 9-year-old male presented with developmental delay, gait abnormalities, seizures, macrocephaly, dysarthria, spasticity, and hyperreflexia. MRI revealed subcortical cysts with diffuse cerebral white matter involvement. Whole-exome sequencing (WES) in the proband did not reveal any clinically relevant single nucleotide variants. However, copy number variation analysis from the WES data of the proband revealed a copy number of 4 for exons 3 and 4 of HEPACAM. Validation and segregation were done by quantitative PCR which confirmed the homozygous duplication of these exons in the proband and carrier status in both parents. To the best of our knowledge, this is the first report of an intragenic duplication in HEPACAM causing MLC2A.
Collapse
Affiliation(s)
- Namanpreet Kaur
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Khyati Arora
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Periyasamy Radhakrishnan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Dhanya Lakshmi Narayanan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
- DBT-Wellcome Trust India Alliance Early Career Clinical and Public Health Research Fellow, Hyderabad, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
8
|
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
|
9
|
Zha J, Chen Y, Cao F, Xu Y, Yang Z, Wen S, Liang M, Wu H, Zhong J. Homozygous variant of MLC1 results in megalencephalic leukoencephalopathy with subcortical cysts. Mol Genet Genomic Med 2024; 12:e2394. [PMID: 38337154 PMCID: PMC10858299 DOI: 10.1002/mgg3.2394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare, inherited disorder that causes epilepsy, intellectual disorders, and early onset macrocephaly. MLC1 has been identified as a main pathogenic gene. METHODS Clinical data such as magnetic resonance imaging (MRI), routine blood tests, and physical examinations were collected from proband. Trio whole-exome sequencing (WES) of the family was performed, and all variants with a minor allele frequency (<0.01) in the exon and canonical splicing sites were selected for further pathogenic evaluation. Candidate variants were validated using Sanger sequencing. RESULTS Here, we report a new homozygous variant identified in two children from the same family in the MLC1 gene [NM_015166.4: c.838_843delinsATTTTA, (p.Ser280_Phe281delinsIleLeu)]. This variant is classified as variant of uncertain significance (VUS) according to the ACMG guidelines. Further experiments demonstrate that the newly identified variant causes a decrease of MLC1 protein levels when expressed in a heterologous expression system. CONCLUSION Our case expands on this genetic variation and provides new evidence for the clinical diagnosis of MLC1-related MLC.
Collapse
Affiliation(s)
- Jian Zha
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| | - Yong Chen
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| | - Fangfang Cao
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| | - Yuxin Xu
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| | | | | | | | - Huaping Wu
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| | - Jianmin Zhong
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangJiangxiChina
| |
Collapse
|
10
|
Kameyama T, Miyata M, Shiotani H, Adachi J, Kakuta S, Uchiyama Y, Mizutani K, Takai Y. Heterogeneity of perivascular astrocyte endfeet depending on vascular regions in the mouse brain. iScience 2023; 26:108010. [PMID: 37829206 PMCID: PMC10565786 DOI: 10.1016/j.isci.2023.108010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Astrocytes interact with not only synapses but also brain blood vessels through perivascular astrocyte endfeet (PV-AEF) to form the neurovascular unit (NVU). However, PV-AEF components have not been fully identified. Here, we biochemically isolated blood vessels from mouse brain homogenates and purified PV-AEF. The purified PV-AEF were observed in different sizes, similar to PV-AEF on brain blood vessels. Mass spectrometry analysis identified 9,762 proteins in the purified PV-AEF, including cell adhesion molecules, nectin-2δ, Kirrel2, and podoplanin. Immunofluorescence microscopic analysis revealed that nectin-2δ and podoplanin were concentrated mainly in arteries/arterioles and veins/venules of the mouse brain, whereas Kirrel2 was mainly in arteries/arterioles. Nectin-2α/δ, Kirrel2, and podoplanin were preferentially observed in large sizes of the purified PV-AEF. Furthermore, Kirrel2 potentially has cell adhesion activity of cultured astrocytes. Collectively, these results indicate that PV-AEF have heterogeneity in sizes and molecular components, implying different roles of PV-AEF in NVU function depending on vascular regions.
Collapse
Affiliation(s)
- Takeshi Kameyama
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima 770-8503, Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yasuo Uchiyama
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| |
Collapse
|
11
|
Chen X, Qu H, Yao Q, Cai X, He T, Zhang X. Case report: Analysis of a gene variant and prenatal diagnosis in a family with megalencephalic leukoencephalopathy with subcortical cysts. Front Neurol 2023; 14:1253398. [PMID: 37928140 PMCID: PMC10622957 DOI: 10.3389/fneur.2023.1253398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare inherited cerebral white matter disorder in children. Pathogenic variations in the causative gene MLC1 are found in approximately 76% of patients and are inherited in an autosomal recessive manner. In this study, we identified an IVS2 + 1delG variant in MLC1 in the firstborn girl of a pregnant woman who has the clinical features of MLC, including macrocephaly, motor development delay, progressive functional deterioration, and myelinopathy, whereas no obvious subcortical cysts were observed by magnetic resonance imaging of the brain. The proband is homozygous for the IVS2 + 1delG mutation, which was inherited from the parents. This variant disrupts the donor splice site, causing an abnormal transcript that results in a premature termination codon and produces a truncated protein, which was confirmed to affect splicing by MLC1 cDNA analysis. This variant was also detected in family members, and a prenatal diagnosis for the fetus was undertaken. Eventually, the couple gave birth to an unaffected baby. Furthermore, we conducted a long-term follow-up of the proband's clinical course. This report improves our understanding of the genetic and phenotypic characteristics of MLC and provides a new genetic basis for prenatal diagnosis and genetic counseling.
Collapse
Affiliation(s)
- Xi Chen
- Department of Medical Genetics and Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Haibo Qu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qiang Yao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaotang Cai
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Department of Rehabilitation, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tiantian He
- Department of Medical Genetics and Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xuemei Zhang
- Department of Medical Genetics and Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
12
|
Das S, Ray BK, Chakraborty U, Tippabathani J, Santra A. The Coexistence of Two Genetic Astrocytopathies-Megalencephalic Leukoencephalopathy and Vanishing White Matter Disease-in an Indian Child. Neuropediatrics 2023; 54:153-156. [PMID: 36878222 DOI: 10.1055/s-0042-1760367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
A 9-month-old male child, born of second-degree consanguinity, presented with a progressively enlarging head since early infancy. The child had normal early development, but further acquisition of milestones after 6 months was delayed. He had afebrile seizures at 9 months, followed by the appearance of appendicular spasticity. First magnetic resonance imaging (MRI) showed nonenhancing, diffuse, bilaterally symmetrical T1/fluid-attenuated inversion recovery (FLAIR) hypointensity and T2 hyperintensity of the cerebral white matter and anterior temporal cysts. Subsequently, the periventricular and deep white matter developed microcystic changes with a pattern of radial stripes. Next-generation sequencing revealed homozygous autosomal recessive variations in the MLC1 gene [c.188T > G, (p.Leu63Arg)] on exon 3 and also in the EIF2B3 gene [c.674G > A, (p.Arg225Gln)] on exon 7, the parents being heterozygous carriers for both variations. This article highlights the rare occurrence of two leukodystrophies of diverse pathogenesis in a child from a nonpredisposed community.
Collapse
Affiliation(s)
- Suman Das
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Biman Kanti Ray
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | - Uddalak Chakraborty
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| | | | - Arindam Santra
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, West Bengal, India
| |
Collapse
|
13
|
Tabata H. Crosstalk between Blood Vessels and Glia during the Central Nervous System Development. Life (Basel) 2022; 12:1761. [PMID: 36362915 PMCID: PMC9699316 DOI: 10.3390/life12111761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/07/2023] Open
Abstract
The formation of proper blood vessel patterns in the central nervous system (CNS) is crucial to deliver oxygen and nutrient to neurons efficiently. At the same time, neurons must be isolated from the outer blood circulation by a specialized structure, the blood-brain barrier (BBB), to maintain the microenvironment of brain parenchyma for the survival of neurons and proper synaptic transmission. To develop this highly organized structure, glial cells, a major component of the brain, have been reported to play essential roles. In this review, the crosstalk between the macroglia, including astrocytes and oligodendrocytes, and endothelial cells during the development of CNS will be discussed. First, the known roles of astrocytes in neuro-vascular unit and its development, and then, the requirements of astrocytes for BBB development and maintenance are shown. Then, various genetic and cellular studies revealing the roles of astrocytes in the growth of blood vessels by providing a scaffold, including laminins and fibronectin, as well as by secreting trophic factors, including vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) are introduced. Finally, the interactions between oligodendrocyte progenitors and blood vessels are overviewed. Although these studies revealed the necessity for proper communication between glia and endothelial cells for CNS development, our knowledge about the detailed cellular and molecular mechanisms for them is still limited. The questions to be clarified in the future are also discussed.
Collapse
Affiliation(s)
- Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| |
Collapse
|
14
|
Perfilyeva A, Bespalova K, Perfilyeva Y, Skvortsova L, Musralina L, Zhunussova G, Khussainova E, Iskakova U, Bekmanov B, Djansugurova L. Integrative Functional Genomic Analysis in Multiplex Autism Families from Kazakhstan. DISEASE MARKERS 2022; 2022:1509994. [PMID: 36199823 PMCID: PMC9529466 DOI: 10.1155/2022/1509994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 12/14/2022]
Abstract
The study of extended pedigrees containing autism spectrum disorder- (ASD-) related broader autism phenotypes (BAP) offers a promising approach to the search for ASD candidate variants. Here, a total of 650,000 genetic markers were tested in four Kazakhstani multiplex families with ASD and BAP to obtain data on de novo mutations (DNMs), common, and rare inherited variants that may contribute to the genetic risk for developing autistic traits. The variants were analyzed in the context of gene networks and pathways. Several previously well-described enriched pathways were identified, including ion channel activity, regulation of synaptic function, and membrane depolarization. Perhaps these pathways are crucial not only for the development of ASD but also for ВАР. The results also point to several additional biological pathways (circadian entrainment, NCAM and BTN family interactions, and interaction between L1 and Ankyrins) and hub genes (CFTR, NOD2, PPP2R2B, and TTR). The obtained results suggest that further exploration of PPI networks combining ASD and BAP risk genes can be used to identify novel or overlooked ASD molecular mechanisms.
Collapse
Affiliation(s)
| | - Kira Bespalova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
- Al-Farabi Kazakh National University, 71 Al-Farabi Ave., Almaty 050040, Kazakhstan
| | - Yuliya Perfilyeva
- M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty 050012, Kazakhstan
- Branch of the National Center for Biotechnology, 14 Zhahanger St., Almaty 050054, Kazakhstan
| | - Liliya Skvortsova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Lyazzat Musralina
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Gulnur Zhunussova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Elmira Khussainova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Ulzhan Iskakova
- Kazakh National Medical University, 94 Tole Bi St., Almaty 050000, Kazakhstan
| | - Bakhytzhan Bekmanov
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| | - Leyla Djansugurova
- Institute of Genetics and Physiology, 93 Al-Farabi Ave., Almaty 050060, Kazakhstan
| |
Collapse
|
15
|
Khalaf-Nazzal R, Dweikat I, Maree M, Alawneh M, Barahmeh M, Doulani RT, Qrareya M, Qadi M, Dudin A. Prevalent MLC1 mutation causing autosomal recessive megalencephalic leukoencephalopathy in consanguineous Palestinian families. Brain Dev 2022; 44:454-461. [PMID: 35440380 DOI: 10.1016/j.braindev.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Recessive forms of megalencephalic leukoencephalopathy with subcortical cysts (MLC, OMIM 604004) is a rare early-onset leukodystrophy that presents with macrocephaly, seizures, slowly progressive gross motor deterioration, and MRI evidence of diffuse symmetric white matter swelling and subcortical cysts in the anterior temporal and frontoparietal regions. Later in the disease course, significant spasticity and ataxia develop, which may be accompanied by intellectual deterioration. This disease is caused mostly by biallelic pathogenic variants in the MLC1 gene. METHODS In this study, we analysed the clinical and molecular architecture of 6 individuals, belonging to 4 unrelated consanguineous Palestinian families, presenting with consistent MLC features. We sequenced the entire coding and flanking intronic regions of the MLC1 gene. RESULTS In all recruited individuals, we detected one recurrent homozygous splice donor mutation NM_015166.4: c.423 + 1G > A. All parents were heterozygous carriers. The mutation abolishes a highly conserved splice site in humans and other species. In silico splice predictors suggested the loss of a canonical splice donor site (CADD score 33.0. SpliceAI: 0.980). The c.423 + 1G > A variant is rare; it was detected in only 4 heterozygous carriers in gnomAD. CONCLUSION In this study, we identified a recurrent MLC1 variant (c.423 + 1G > A) as the cause of MLC among a group of Palestinian patients originating from a particular region of the country. Cost-effective studies should be performed to evaluate the implementation of carrier screening in adults originating from this region. Our findings have the potential to contribute to improved genetic diagnosis and carrier testing for individuals within this population and the wider community.
Collapse
Affiliation(s)
- Reham Khalaf-Nazzal
- Faculty of Medicine, Arab American University of Palestine, Jenin, Palestine.
| | - Imad Dweikat
- Faculty of Medicine, Arab American University of Palestine, Jenin, Palestine
| | - Mosab Maree
- Medicine Department, Faculty of Medicine and Health Sciences, An-Najah National University, and An-Najah National University Hospital, Nablus, Palestine
| | - Maysa Alawneh
- Medicine Department, Faculty of Medicine and Health Sciences, An-Najah National University, and An-Najah National University Hospital, Nablus, Palestine
| | - Myassar Barahmeh
- Biomedical Sciences Department, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Rasha T Doulani
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Qrareya
- Clinical Sciences Department, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Qadi
- Biomedical Sciences Department, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Anwar Dudin
- Pediatric Neurology Outpatient Clinic, Ramallah, Palestine
| |
Collapse
|
16
|
Schrader JM, Xu F, Lee H, Barlock B, Benveniste H, Van Nostrand WE. Emergent White Matter Degeneration in the rTg-DI Rat Model of Cerebral Amyloid Angiopathy Exhibits Unique Proteomic Changes. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:426-440. [PMID: 34896071 PMCID: PMC8895424 DOI: 10.1016/j.ajpath.2021.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022]
Abstract
Cerebral amyloid angiopathy (CAA), characterized by cerebral vascular amyloid accumulation, neuroinflammation, microbleeds, and white matter (WM) degeneration, is a common comorbidity in Alzheimer disease and a prominent contributor to vascular cognitive impairment and dementia. WM loss was recently reported in the corpus callosum (CC) in the rTg-DI rat model of CAA. The current study shows that the CC exhibits a much lower CAA burden compared with the adjacent cortex. Sequential Window Acquisition of All Theoretical Mass Spectra tandem mass spectrometry was used to show specific proteomic changes in the CC with emerging WM loss and compare them with the proteome of adjacent cortical tissue in rTg-DI rats. In the CC, annexin A3, heat shock protein β1, and cystatin C were elevated at 4 months (M) before WM loss and at 12M with evident WM loss. Although annexin A3 and cystatin C were also enhanced in the cortex at 12M, annexin A5 and the leukodystrophy-associated astrocyte proteins megalencephalic leukoencephalopathy with subcortical cysts 1 and GlialCAM were distinctly elevated in the CC. Pathway analysis indicated neurodegeneration of axons, reflected by reduced expression of myelin and neurofilament proteins, was common to the CC and cortex; activation of Tgf-β1 and F2/thrombin was restricted to the CC. This study provides new insights into the proteomic changes that accompany WM loss in the CC of rTg-DI rats.
Collapse
Affiliation(s)
- Joseph M Schrader
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Hedok Lee
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - Benjamin Barlock
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Helene Benveniste
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - William E Van Nostrand
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island.
| |
Collapse
|
17
|
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: 0.5] [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
|
18
|
Ain Ul Batool S, Almatrafi A, Fadhli F, Alluqmani M, Ali G, Basit S. A homozygous missense variant in the MLC1 gene underlies megalencephalic leukoencephalopathy with subcortical cysts in large kindred: Heterozygous carriers show seizure and mild motor function deterioration. Am J Med Genet A 2021; 188:1075-1082. [PMID: 34918859 DOI: 10.1002/ajmg.a.62614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/17/2021] [Accepted: 11/28/2021] [Indexed: 11/07/2022]
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy characterized by epileptic seizures, macrocephaly, and vacuolization of myelin and astrocyte. The magnetic resonance imaging of the brain of MLC patients shows diffuse white-matter anomalies and the occurrence of subcortical cysts. MLC features have been observed in individuals having mutations in the MLC1 or HEPACAM genes. In this study, we recruited a six generation large kindred with five affected individuals manifesting clinical features of epileptic seizures, macrocephaly, ataxia, and spasticity. In order to identify the underlying genetic cause of the clinical features, we performed whole-genome genotyping using Illumina microarray followed by detection of loss of heterozygosity (LOHs) regions. One affected individual was exome sequenced as well. Homozygosity mapping detected several LOH regions due to extensive consanguinity. An unbiased and hypothesis-free exome data analysis identified a homozygous missense variant (NM_015166.3:c.278C>T) in the exon 4 of the MLC1 gene. The variant is present in the LOH region on chromosome 22q (50 Mb) and segregates perfectly with the disorder within the family in an autosomal recessive manner. The variant is present in a highly conserved first cytoplasmic domain of the MLC1 protein (NM_015166.3:p.(Ser93Leu)). Interestingly, heterozygous individuals show seizure and mild motor function deterioration. We propose that the heterozygous variant in MLC1 might disrupt the functional interaction of MLC1 with GlialCAM resulting in mild clinical features in carriers of the variant.
Collapse
Affiliation(s)
- Syeda Ain Ul Batool
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Ahmad Almatrafi
- Department of Biology, College of Science, Taibah University, Medina, Saudi Arabia
| | - Fatima Fadhli
- Department of Genetics, Madinah Maternity and Children Hospital, Medina, Saudi Arabia
| | - Majed Alluqmani
- Department of Neurology, College of Medicine, Taibah University Medina, Saudi Arabia
| | - Ghazanfar Ali
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University Medina, Medina, Saudi Arabia
| |
Collapse
|
19
|
Hwang J, Park K, Lee GY, Yoon BY, Kim H, Roh SH, Lee BC, Kim K, Lim HH. Transmembrane topology and oligomeric nature of an astrocytic membrane protein, MLC1. Open Biol 2021; 11:210103. [PMID: 34847774 PMCID: PMC8633789 DOI: 10.1098/rsob.210103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
MLC1 is a membrane protein mainly expressed in astrocytes, and genetic mutations lead to the development of a leukodystrophy, megalencephalic leukoencephalopathy with subcortical cysts disease. Currently, the biochemical properties of the MLC1 protein are largely unknown. In this study, we aimed to characterize the transmembrane (TM) topology and oligomeric nature of the MLC1 protein. Systematic immunofluorescence staining data revealed that the MLC1 protein has eight TM domains and that both the N- and C-terminus face the cytoplasm. We found that MLC1 can be purified as an oligomer and could form a trimeric complex in both detergent micelles and reconstituted proteoliposomes. Additionally, a single-molecule photobleaching experiment showed that MLC1 protein complexes could consist of three MLC1 monomers in the reconstituted proteoliposomes. These results can provide a basis for both the high-resolution structural determination and functional characterization of the MLC1 protein.
Collapse
Affiliation(s)
- Junmo Hwang
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu 41068, Republic of Korea
| | - Kunwoong Park
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu 41068, Republic of Korea
| | - Ga-Young Lee
- Brain Research Core Facility, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Bo Young Yoon
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu 41068, Republic of Korea
| | - Hyunmin Kim
- School of Biological Science, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Sung Hoon Roh
- School of Biological Science, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Byoung-Cheol Lee
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu 41068, Republic of Korea
| | - Kipom Kim
- Brain Research Core Facility, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu 41068, Republic of Korea,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| |
Collapse
|
20
|
Control of membrane protein homeostasis by a chaperone-like glial cell adhesion molecule at multiple subcellular locations. Sci Rep 2021; 11:18435. [PMID: 34531445 PMCID: PMC8446001 DOI: 10.1038/s41598-021-97777-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/24/2021] [Indexed: 01/17/2023] Open
Abstract
The significance of crosstalks among constituents of plasma membrane protein clusters/complexes in cellular proteostasis and protein quality control (PQC) remains incompletely understood. Examining the glial (enriched) cell adhesion molecule (CAM), we demonstrate its chaperone-like role in the biosynthetic processing of the megalencephalic leukoencephalopathy with subcortical cyst 1 (MLC1)-heteromeric regulatory membrane protein complex, as well as the function of the GlialCAM/MLC1 signalling complex. We show that in the absence of GlialCAM, newly synthesized MLC1 molecules remain unfolded and are susceptible to polyubiquitination-dependent proteasomal degradation at the endoplasmic reticulum. At the plasma membrane, GlialCAM regulates the diffusional partitioning and endocytic dynamics of cluster members, including the ClC-2 chloride channel and MLC1. Impaired folding and/or expression of GlialCAM or MLC1 in the presence of diseases causing mutations, as well as plasma membrane tethering compromise the functional expression of the cluster, leading to compromised endo-lysosomal organellar identity. In addition, the enlarged endo-lysosomal compartments display accelerated acidification, ubiquitinated cargo-sorting and impaired endosomal recycling. Jointly, these observations indicate an essential and previously unrecognized role for CAM, where GliaCAM functions as a PQC factor for the MLC1 signalling complex biogenesis and possess a permissive role in the membrane dynamic and cargo sorting functions with implications in modulations of receptor signalling.
Collapse
|
21
|
Amin M, Vignal C, Hamed AAA, Mohammed IN, Elseed MA, Drunat S, Babai A, Eltaraifee E, Elbadi I, Abubaker R, Mustafa D, Yahia A, Koko M, Osman M, Bakhit Y, Elshafea A, Alsiddig M, Haroun S, Lelay G, Elsayed LEO, Ahmed AE, Boespflug-Tanguy O, Dorboz I. Novel variants causing megalencephalic leukodystrophy in Sudanese families. J Hum Genet 2021; 67:127-132. [PMID: 34504271 DOI: 10.1038/s10038-021-00945-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/12/2021] [Accepted: 05/28/2021] [Indexed: 11/09/2022]
Abstract
Mutations in MLC1 cause megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare form of leukodystrophy characterized by macrocephaly, epilepsy, spasticity, and slow mental deterioration. Genetic studies of MLC are lacking from many parts of the world, especially in Sub-Saharan Africa. Genomic DNA was extracted for 67 leukodystrophic patients from 43 Sudanese families. Mutations were screened using the NGS panel testing 139 leukodystrophies and leukoencephalopathies causing genes (NextSeq500 Illumina). Five homozygous MLC1 variants were discovered in seven patients from five distinct families, including three consanguineous families from the same region of Sudan. Three variants were missense (c.971 T > G, p.Ile324Ser; c.344 T > C, p.Phe115Ser; and c.881 C > T, p.Pro294Leu), one duplication (c.831_838dupATATCTGT, p.Ser280Tyrfs*8), and one synonymous/splicing-site mutation (c.762 C > T, p.Ser254). The segregation pattern was consistent with autosomal recessive inheritance. The clinical presentation and brain MRI of the seven affected patients were consistent with the diagnosis of MLC1. Due to the high frequency of distinct MLC1 mutations found in our leukodystrophic Sudanese families, we analyzed the coding sequence of MLC1 gene in 124 individuals from the Sudanese genome project in comparison with the 1000-genome project. We found that Sudan has the highest proportion of deleterious variants in MLC1 gene compared with other populations from the 1000-genome project.
Collapse
Affiliation(s)
- Mutaz Amin
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan.,Université de Paris, NeuroDiderot, UMR 1141, INSERM, Paris, France
| | - Cedric Vignal
- Unité de Génétique Moleculaire, Departement de Genetique Médicale, APHP, Hopital Robert-Debré, Paris, France
| | - Ahlam A A Hamed
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Maha A Elseed
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Severine Drunat
- Université de Paris, NeuroDiderot, UMR 1141, INSERM, Paris, France.,Unité de Génétique Moleculaire, Departement de Genetique Médicale, APHP, Hopital Robert-Debré, Paris, France
| | - Arwa Babai
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Iman Elbadi
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Rayan Abubaker
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Doaa Mustafa
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Ashraf Yahia
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan.,Institut du Cerveau, INSERM U1127, CNRS UMR7225, Sorbonne Université, Paris, France
| | - Mahmoud Koko
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Melka Osman
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Yousuf Bakhit
- Faculty of Dentistry, University of Khartoum, Khartoum, Sudan
| | - Azza Elshafea
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | - Sahwah Haroun
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Gurvan Lelay
- Université de Paris, NeuroDiderot, UMR 1141, INSERM, Paris, France
| | | | - Ammar E Ahmed
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Odile Boespflug-Tanguy
- Université de Paris, NeuroDiderot, UMR 1141, INSERM, Paris, France.,Neuropediatrie, LEUKOFRANCE, APHP, Hopital Robert-Debré, Paris, France
| | - Imen Dorboz
- Université de Paris, NeuroDiderot, UMR 1141, INSERM, Paris, France.
| |
Collapse
|
22
|
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: 9] [Impact Index Per Article: 2.3] [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
|
23
|
Elorza-Vidal X, Xicoy-Espaulella E, Pla-Casillanis A, Alonso-Gardón M, Gaitán-Peñas H, Engel-Pizcueta C, Fernández-Recio J, Estévez R. Structural basis for the dominant or recessive character of GLIALCAM mutations found in leukodystrophies. Hum Mol Genet 2021; 29:1107-1120. [PMID: 31960914 PMCID: PMC7206653 DOI: 10.1093/hmg/ddaa009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a type of leukodystrophy characterized by white matter edema, and it is caused mainly by recessive mutations in MLC1 and GLIALCAM genes. These variants are called MLC1 and MLC2A with both types of patients sharing the same clinical phenotype. In addition, dominant mutations in GLIALCAM have also been identified in a subtype of MLC patients with a remitting phenotype. This variant has been named MLC2B. GLIALCAM encodes for an adhesion protein containing two immunoglobulin (Ig) domains and it is needed for MLC1 targeting to astrocyte–astrocyte junctions. Most mutations identified in GLIALCAM abolish GlialCAM targeting to junctions. However, it is unclear why some mutations behave as recessive or dominant. Here, we used a combination of biochemistry methods with a new developed anti-GlialCAM nanobody, double-mutants and cysteine cross-links experiments, together with computer docking, to create a structural model of GlialCAM homo-interactions. Using this model, we suggest that dominant mutations affect different GlialCAM–GlialCAM interacting surfaces in the first Ig domain, which can occur between GlialCAM molecules present in the same cell (cis) or present in neighbouring cells (trans). Our results provide a framework that can be used to understand the molecular basis of pathogenesis of all identified GLIALCAM mutations.
Collapse
Affiliation(s)
- 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, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| | - Efren Xicoy-Espaulella
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Adrià Pla-Casillanis
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Marta Alonso-Gardón
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Héctor Gaitán-Peñas
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| | - Carolyn Engel-Pizcueta
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Juan Fernández-Recio
- Barcelona Supercomputing Center (BSC), Barcelona, Spain.,Institut de Biologia Molecular de Barcelona, CSIC, Barcelona, Spain.,Instituto de Ciencias de la Vid y del Vino (ICVV), CSIC- Universidad de La Rioja- Gobierno de la Rioja, Logroño, 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, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| |
Collapse
|
24
|
Baldwin KT, Tan CX, Strader ST, Jiang C, Savage JT, Elorza-Vidal X, Contreras X, Rülicke T, Hippenmeyer S, Estévez R, Ji RR, Eroglu C. HepaCAM controls astrocyte self-organization and coupling. Neuron 2021; 109:2427-2442.e10. [PMID: 34171291 PMCID: PMC8547372 DOI: 10.1016/j.neuron.2021.05.025] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 04/19/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
Astrocytes extensively infiltrate the neuropil to regulate critical aspects of synaptic development and function. This process is regulated by transcellular interactions between astrocytes and neurons via cell adhesion molecules. How astrocytes coordinate developmental processes among one another to parse out the synaptic neuropil and form non-overlapping territories is unknown. Here we identify a molecular mechanism regulating astrocyte-astrocyte interactions during development to coordinate astrocyte morphogenesis and gap junction coupling. We show that hepaCAM, a disease-linked, astrocyte-enriched cell adhesion molecule, regulates astrocyte competition for territory and morphological complexity in the developing mouse cortex. Furthermore, conditional deletion of Hepacam from developing astrocytes significantly impairs gap junction coupling between astrocytes and disrupts the balance between synaptic excitation and inhibition. Mutations in HEPACAM cause megalencephalic leukoencephalopathy with subcortical cysts in humans. Therefore, our findings suggest that disruption of astrocyte self-organization mechanisms could be an underlying cause of neural pathology.
Collapse
Affiliation(s)
- Katherine T Baldwin
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Christabel X Tan
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Samuel T Strader
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Changyu Jiang
- Department of Anesthesiology and Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Justin T Savage
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xabier Elorza-Vidal
- Unitat de Fisiología, Departament de Ciències Fisiològiques, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Ximena Contreras
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Raúl Estévez
- Unitat de Fisiología, Departament de Ciències Fisiològiques, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Ru-Rong Ji
- Department of Anesthesiology and Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences (DIBS), Durham, NC 27710, USA; Duke University Regeneration Next Initiative, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Ravi K, Paidas MJ, Saad A, Jayakumar AR. Astrocytes in rare neurological conditions: Morphological and functional considerations. J Comp Neurol 2021; 529:2676-2705. [PMID: 33496339 DOI: 10.1002/cne.25118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 01/06/2023]
Abstract
Astrocytes are a population of central nervous system (CNS) cells with distinctive morphological and functional characteristics that differ within specific areas of the brain and are widely distributed throughout the CNS. There are mainly two types of astrocytes, protoplasmic and fibrous, which differ in morphologic appearance and location. Astrocytes are important cells of the CNS that not only provide structural support, but also modulate synaptic activity, regulate neuroinflammatory responses, maintain the blood-brain barrier, and supply energy to neurons. As a result, astrocytic disruption can lead to widespread detrimental effects and can contribute to the pathophysiology of several neurological conditions. The characteristics of astrocytes in more common neuropathologies such as Alzheimer's and Parkinson's disease have significantly been described and continue to be widely studied. However, there still exist numerous rare neurological conditions in which astrocytic involvement is unknown and needs to be explored. Accordingly, this review will summarize functional and morphological changes of astrocytes in various rare neurological conditions based on current knowledge thus far and highlight remaining neuropathologies where astrocytic involvement has yet to be investigated.
Collapse
Affiliation(s)
- Karthik Ravi
- University of Michigan, Ann Arbor, Michigan, USA
| | - Michael J Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA
| | - Ali Saad
- Pathology and Laboratory Medicine, University of Miami School of Medicine, Miami, Florida, USA
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami School of Medicine, Miami, Florida, USA.,South Florida VA Foundation for Research and Education Inc, Miami, Florida, USA.,General Medical Research Neuropathology Section, R&D Service, Veterans Affairs Medical Centre, Miami, Florida, USA
| |
Collapse
|
26
|
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: 2.8] [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
|
27
|
Fernandez-Abascal J, Graziano B, Encalada N, Bianchi L. Glial Chloride Channels in the Function of the Nervous System Across Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:195-223. [PMID: 35138616 PMCID: PMC11247392 DOI: 10.1007/978-981-16-4254-8_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the nervous system, the concentration of Cl- in neurons that express GABA receptors plays a key role in establishing whether these neurons are excitatory, mostly during early development, or inhibitory. Thus, much attention has been dedicated to understanding how neurons regulate their intracellular Cl- concentration. However, regulation of the extracellular Cl- concentration by other cells of the nervous system, including glia and microglia, is as important because it ultimately affects the Cl- equilibrium potential across the neuronal plasma membrane. Moreover, Cl- ions are transported in and out of the cell, via either passive or active transporter systems, as counter ions for K+ whose concentration in the extracellular environment of the nervous system is tightly regulated because it directly affects neuronal excitability. In this book chapter, we report on the Cl- channel types expressed in the various types of glial cells focusing on the role they play in the function of the nervous system in health and disease. Furthermore, we describe the types of stimuli that these channels are activated by, the other solutes that they may transport, and the involvement of these channels in processes such as pH regulation and Regulatory Volume Decrease (RVD). The picture that emerges is one of the glial cells expressing a variety of Cl- channels, encoded by members of different gene families, involved both in short- and long-term regulation of the nervous system function. Finally, we report data on invertebrate model organisms, such as C. elegans and Drosophila, that are revealing important and previously unsuspected functions of some of these channels in the context of living and behaving animals.
Collapse
Affiliation(s)
- Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Bianca Graziano
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Nicole Encalada
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
28
|
Lattier JM, De A, Chen Z, Morales JE, Lang FF, Huse JT, McCarty JH. Megalencephalic leukoencephalopathy with subcortical cysts 1 (MLC1) promotes glioblastoma cell invasion in the brain microenvironment. Oncogene 2020; 39:7253-7264. [PMID: 33040087 PMCID: PMC7736299 DOI: 10.1038/s41388-020-01503-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/17/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM), or grade IV astrocytoma, is a malignant brain cancer that contains subpopulations of proliferative and invasive cells that coordinately drive primary tumor growth, progression, and recurrence after therapy. Here, we have analyzed functions for megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1), an eight-transmembrane protein normally expressed in perivascular brain astrocyte end feet that is essential for neurovascular development and physiology, in the pathogenesis of GBM. We show that Mlc1 is expressed in human stem-like GBM cells (GSCs) and is linked to the development of primary and recurrent GBM. Genetically inhibiting MLC1 in GSCs using RNAi-mediated gene silencing results in diminished growth and invasion in vitro as well as impaired tumor initiation and progression in vivo. Biochemical assays identify the receptor tyrosine kinase Axl and its intracellular signaling effectors as important for MLC1 control of GSC invasive growth. Collectively, these data reveal key functions for MLC1 in promoting GSC growth and invasion, and suggest that targeting the Mlc1 protein or its associated signaling effectors may be a useful therapy for blocking tumor progression in patients with primary or recurrent GBM.
Collapse
Affiliation(s)
- John M Lattier
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Arpan De
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Zhihua Chen
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - John E Morales
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Frederick F Lang
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jason T Huse
- Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Joseph H McCarty
- Departments of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
de Waard DM, Bugiani M. Astrocyte-Oligodendrocyte-Microglia Crosstalk in Astrocytopathies. Front Cell Neurosci 2020; 14:608073. [PMID: 33328899 PMCID: PMC7710860 DOI: 10.3389/fncel.2020.608073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Defective astrocyte function due to a genetic mutation can have major consequences for microglia and oligodendrocyte physiology, which in turn affects the white matter integrity of the brain. This review addresses the current knowledge on shared and unique pathophysiological mechanisms of astrocytopathies, including vanishing white matter, Alexander disease, megalencephalic leukoencephalopathy with subcortical cysts, Aicardi-Goutières syndrome, and oculodentodigital dysplasia. The mechanisms of disease include protein accumulation, unbalanced secretion of extracellular matrix proteins, pro- and anti-inflammatory molecules, cytokines and chemokines by astrocytes, as well as an altered gap junctional network and a changed ionic and nutrient homeostasis. Interestingly, the extent to which astrogliosis and microgliosis are present in these astrocytopathies is highly variable. An improved understanding of astrocyte-microglia-oligodendrocyte crosstalk might ultimately lead to the identification of druggable targets for these, currently untreatable, severe conditions.
Collapse
Affiliation(s)
| | - Marianna Bugiani
- Department of Pathology, VU Medical center, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
30
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- 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, France
| | - Leila Slaoui
- 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, France
| | - Noémie Mazaré
- 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, France
| | - 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, France
| | - Marc Oudart
- 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, France
| | - Rodrigo Alvear-Perez
- 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, France
| | - Xabier Elorza-Vidal
- 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, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- 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, France
| |
Collapse
|
31
|
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: 7] [Impact Index Per Article: 1.4] [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
|
32
|
Fujino S, Yoshihashi H, Takeda R, Ihara S, Miyama S. White matter abnormality in Jacobsen syndrome assessed by serial MRI. Brain Dev 2020; 42:621-625. [PMID: 32507665 DOI: 10.1016/j.braindev.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/09/2020] [Accepted: 05/03/2020] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Jacobsen syndrome (JS) is caused by a deletion at the terminus of the long arm of chromosome 11. There are few reports of JS associated with cerebral white matter abnormalities (WMA), and the etiology, pathophysiology, and time-dependent changes in WMA with JS still remain unclear. CASE REPORT The patient was a 2-month-old female with several morphological anomalies, including trigonocephaly, ectropion, flat nasal bridge, low-set ears, and sparse eyebrows. Chromosome analysis (G-banding karyotyping) of 46,XX,del(11)(q23.3) led to the diagnosis of JS. Head MRI performed at age 9 months indicated diffuse WMA with hyperintense signals on T2-weighted imaging. MRI at age 2.5 years demonstrated a decrease in the WMA and progressive myelination. DISCUSSION These findings suggested that the WMA in the present patient were due to chronic white matter edema associated with a deletion in the 11q terminal region of HEPACAM/GlialCAM, a causative gene for megalencephalic leukoencephalopathy with subcortical cysts type 2B (MLC2B). As with some of MLC2B patients, the WMA in the present patient improved over time. The present report is the first to document dramatic changes in WMA in JS visualized by serial MRI examinations from the neonatal period through early childhood. CONCLUSION The findings of the present study suggested that WMA in JS are due to chronic white matter edema associated with HEPACAM/GlialCAM deletion and show gradual improvement over time, as seen in some MLC2B patients.
Collapse
Affiliation(s)
- Shuhei Fujino
- Department of Neurology, Tokyo Metropolitan Children's Medical Center, Japan.
| | - Hiroshi Yoshihashi
- Department of Medical Genetics, Tokyo Metropolitan Children's Medical Center, Japan
| | - Ryojun Takeda
- Department of Medical Genetics, Tokyo Metropolitan Children's Medical Center, Japan
| | - Satoshi Ihara
- Department of Neurosurgery, Tokyo Metropolitan Children's Medical Center, Japan
| | - Sahoko Miyama
- Department of Neurology, Tokyo Metropolitan Children's Medical Center, Japan
| |
Collapse
|
33
|
Megalencephalic Leukoencephalopathy with Subcortical Cysts Disease-Linked MLC1 Protein Favors Gap-Junction Intercellular Communication by Regulating Connexin 43 Trafficking in Astrocytes. Cells 2020; 9:cells9061425. [PMID: 32521795 PMCID: PMC7348769 DOI: 10.3390/cells9061425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 01/06/2023] Open
Abstract
Astrocytes, the most numerous cells of the central nervous system, exert critical functions for brain homeostasis. To this purpose, astrocytes generate a highly interconnected intercellular network allowing rapid exchange of ions and metabolites through gap junctions, adjoined channels composed of hexamers of connexin (Cx) proteins, mainly Cx43. Functional alterations of Cxs and gap junctions have been observed in several neuroinflammatory/neurodegenerative diseases. In the rare leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC), astrocytes show defective control of ion/fluid exchanges causing brain edema, fluid cysts, and astrocyte/myelin vacuolation. MLC is caused by mutations in MLC1, an astrocyte-specific protein of elusive function, and in GlialCAM, a MLC1 chaperon. Both proteins are highly expressed at perivascular astrocyte end-feet and astrocyte-astrocyte contacts where they interact with zonula occludens-1 (ZO-1) and Cx43 junctional proteins. To investigate the possible role of Cx43 in MLC pathogenesis, we studied Cx43 properties in astrocytoma cells overexpressing wild type (WT) MLC1 or MLC1 carrying pathological mutations. Using biochemical and electrophysiological techniques, we found that WT, but not mutated, MLC1 expression favors intercellular communication by inhibiting extracellular-signal-regulated kinase 1/2 (ERK1/2)-mediated Cx43 phosphorylation and increasing Cx43 gap-junction stability. These data indicate MLC1 regulation of Cx43 in astrocytes and Cx43 involvement in MLC pathogenesis, suggesting potential target pathways for therapeutic interventions.
Collapse
|
34
|
D'Adamo MC, Liantonio A, Conte E, Pessia M, Imbrici P. Ion Channels Involvement in Neurodevelopmental Disorders. Neuroscience 2020; 440:337-359. [PMID: 32473276 DOI: 10.1016/j.neuroscience.2020.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Inherited and sporadic mutations in genes encoding for brain ion channels, affecting membrane expression or biophysical properties, have been associated with neurodevelopmental disorders characterized by epilepsy, cognitive and behavioral deficits with significant phenotypic and genetic heterogeneity. Over the years, the screening of a growing number of patients and the functional characterization of newly identified mutations in ion channels genes allowed to recognize new phenotypes and to widen the clinical spectrum of known diseases. Furthermore, advancements in understanding disease pathogenesis at atomic level or using patient-derived iPSCs and animal models have been pivotal to orient therapeutic intervention and to put the basis for the development of novel pharmacological options for drug-resistant disorders. In this review we will discuss major improvements and critical issues concerning neurodevelopmental disorders caused by dysfunctions in brain sodium, potassium, calcium, chloride and ligand-gated ion channels.
Collapse
Affiliation(s)
- Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | | | - Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Italy
| | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Italy.
| |
Collapse
|
35
|
Pleasure D, Guo F, Chechneva O, Bannerman P, McDonough J, Burns T, Wang Y, Hull V. Pathophysiology and Treatment of Canavan Disease. Neurochem Res 2020; 45:561-565. [PMID: 30535831 PMCID: PMC11131954 DOI: 10.1007/s11064-018-2693-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/28/2023]
Affiliation(s)
- David Pleasure
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA.
- , C/o Shriners Hospital, 2425 Stockton Blvd, Sacramento, CA, 95817, USA.
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| | - Olga Chechneva
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| | - Peter Bannerman
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| | - Jennifer McDonough
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44242, USA
| | - Travis Burns
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| | - Yan Wang
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| | - Vanessa Hull
- Institute for Pediatric Regenerative Research, Shriners Hospitals for Children Northern California and UC Davis School of Medicine, 2425 Stockton Blvd, 95817, Sacramento, CA, USA
| |
Collapse
|
36
|
Hwang J, Vu HM, Kim MS, Lim HH. Plasma membrane localization of MLC1 regulates cellular morphology and motility. Mol Brain 2019; 12:116. [PMID: 31888684 PMCID: PMC6938022 DOI: 10.1186/s13041-019-0540-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
Background Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare form of infantile-onset leukodystrophy. The disorder is caused primarily by mutations of MLC1 that leads to a series of phenotypic outcomes including vacuolation of myelin and astrocytes, subcortical cysts, brain edema, and macrocephaly. Recent studies have indicated that functional interactions among MLC1, GlialCAM, and ClC-2 channels play key roles in the regulation of neuronal, glial and vascular homeostasis. However, the physiological role of MLC1 in cellular homeostatic communication remains poorly understood. In the present study, we investigated the cellular function of MLC1 and its effects on cell–cell interactions. Methods MLC1-dependent cellular morphology and motility were analyzed by using confocal and live cell imaging technique. Biochemical approaches such as immunoblotting, co-immunoprecipitation, and surface biotinylation were conducted to support data. Results We found that the altered MLC1 expression and localization led to a great alteration in cellular morphology and motility through actin remodeling. MLC1 overexpression induced filopodia formation and suppressed motility. And, MLC1 proteins expressed in patient-derived MLC1 mutants resulted in trapping in the ER although no changes in morphology or motility were observed. Interestingly knockdown of Mlc1 induced Arp3-Cortactin interaction, lamellipodia formation, and increased the membrane ruffling of the astrocytes. These data indicate that subcellular localization of expressed MLC1 at the plasma membrane is critical for changes in actin dynamics through ARP2/3 complex. Thus, our results suggest that misallocation of pathogenic mutant MLC1 may disturbs the stable cell-cell communication and the homeostatic regulation of astrocytes in patients with MLC.
Collapse
Affiliation(s)
- Junmo Hwang
- Molecular Physiology and Biophysics Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 41062, Daegu, Republic of Korea
| | - Hung M Vu
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 42988, Daegu, Republic of Korea
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 42988, Daegu, Republic of Korea
| | - Hyun-Ho Lim
- Molecular Physiology and Biophysics Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), 41062, Daegu, Republic of Korea. .,Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 42988, Daegu, Republic of Korea.
| |
Collapse
|
37
|
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: 8] [Impact Index Per Article: 1.3] [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
|
38
|
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
|
39
|
Assessment of genetic variant burden in epilepsy-associated brain lesions. Eur J Hum Genet 2019; 27:1738-1744. [PMID: 31358956 DOI: 10.1038/s41431-019-0484-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 06/29/2019] [Accepted: 07/05/2019] [Indexed: 01/31/2023] Open
Abstract
It is challenging to estimate genetic variant burden across different subtypes of epilepsy. Herein, we used a comparative approach to assess the genetic variant burden and genotype-phenotype correlations in four most common brain lesions in patients with drug-resistant focal epilepsy. Targeted sequencing analysis was performed for a panel of 161 genes with a mean coverage of >400×. Lesional tissue was histopathologically reviewed and dissected from hippocampal sclerosis (n = 15), ganglioglioma (n = 16), dysembryoplastic neuroepithelial tumors (n = 8), and focal cortical dysplasia type II (n = 15). Peripheral blood (n = 12) or surgical tissue samples histopathologically classified as lesion-free (n = 42) were available for comparison. Variants were classified as pathogenic or likely pathogenic according to American College of Medical Genetics and Genomics guidelines. Overall, we identified pathogenic and likely pathogenic variants in 25.9% of patients with a mean coverage of 383×. The highest number of pathogenic/likely pathogenic variants was observed in patients with ganglioglioma (43.75%; all somatic) and dysembryoplastic neuroepithelial tumors (37.5%; all somatic), and in 20% of cases with focal cortical dysplasia type II (13.33% somatic, 6.67% germline). Pathogenic/likely pathogenic positive genes were disorder specific and BRAF V600E the only recurrent pathogenic variant. This study represents a reference for the genetic variant burden across the four most common lesion entities in patients with drug-resistant focal epilepsy. The observed large variability in variant burden by epileptic lesion type calls for whole exome sequencing of histopathologically well-characterized tissue in a diagnostic setting and in research to discover novel disease-associated genes.
Collapse
|
40
|
Zanatta AP, Gonçalves R, Zanatta L, de Oliveria GT, Ludwig Moraes AL, Zamoner A, Fernández-Dueñas V, Lanznaster D, Ciruela F, Tasca CI, Delalande C, Menegaz D, Mena Barreto Silva FR. New ionic targets of 3,3′,5′-triiodothyronine at the plasma membrane of rat Sertoli cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:748-759. [DOI: 10.1016/j.bbamem.2019.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 12/18/2018] [Accepted: 01/07/2019] [Indexed: 11/26/2022]
|
41
|
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: 2.8] [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
|
42
|
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: 5.0] [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
|
43
|
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: 3.9] [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
|
44
|
Hamilton EMC, Tekturk P, Cialdella F, van Rappard DF, Wolf NI, Yalcinkaya C, Çetinçelik Ü, Rajaee A, Kariminejad A, Paprocka J, Yapici Z, Bošnjak VM, van der Knaap MS. Megalencephalic leukoencephalopathy with subcortical cysts: Characterization of disease variants. Neurology 2018; 90:e1395-e1403. [PMID: 29661901 PMCID: PMC5902784 DOI: 10.1212/wnl.0000000000005334] [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: 09/14/2017] [Accepted: 01/16/2018] [Indexed: 01/08/2023] Open
Abstract
Objective To provide an overview of clinical and MRI characteristics of the different variants of the leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC) and identify possible differentiating features. Methods We performed an international multi-institutional, cross-sectional observational study of the clinical and MRI characteristics in patients with genetically confirmed MLC. Clinical information was obtained by questionnaires for physicians and retrospective chart review. Results We included 204 patients with classic MLC, 187 of whom had recessive mutations in MLC1 (MLC1 variant) and 17 in GLIALCAM (MLC2A variant) and 38 patients with remitting MLC caused by dominant GLIALCAM mutations (MLC2B variant). We observed a relatively wide variability in neurologic disability among patients with classic MLC. No clinical differences could be identified between patients with MLC1 and MLC2A. Patients with MLC2B invariably had a milder phenotype with preservation of motor function, while intellectual disability and autism were relatively frequent. Systematic MRI review revealed no MRI features that distinguish between MLC1 and MLC2A. Radiologic improvement was observed in all patients with MLC2B and also in 2 patients with MLC1. In MRIs obtained in the early disease stage, absence of signal abnormalities of the posterior limb of the internal capsule and cerebellar white matter and presence of only rarefied subcortical white matter instead of true subcortical cysts were suggestive of MLC2B. Conclusion Clinical and MRI features did not distinguish between classic MLC with MLC1 or GLIALCAM mutations. Absence of signal abnormalities of the internal capsule and cerebellar white matter are MRI findings that point to the remitting phenotype.
Collapse
Affiliation(s)
- Eline M C Hamilton
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Pinar Tekturk
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Fia Cialdella
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Diane F van Rappard
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Nicole I Wolf
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Cengiz Yalcinkaya
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Ümran Çetinçelik
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Ahmad Rajaee
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Ariana Kariminejad
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Justyna Paprocka
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Zuhal Yapici
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Vlatka Mejaški Bošnjak
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Marjo S van der Knaap
- From the Department of Child Neurology and Amsterdam Neuroscience (E.M.C.H., F.C., D.F.v.R., N.I.W., M.S.v.d.K.), VU University Medical Center, Amsterdam, the Netherlands; Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine (P.T., Z.Y.), and Division of Child Neurology, Department of Neurology, Cerrahpasa Medical School (C.Y.), Istanbul University; clinical geneticist in private practice (Ü.Ç.), Merkez Mahallesi, İstanbul, Turkey; Kariminejad-Najmabadi Pathology & Genetics Center (A.R., A.K.), Tehran, Iran; Department of Pediatric Neurology (J.P.), School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland; Department of Neuropediatrics (V.M.B.), Children's Hospital Zagreb, School of Medicine, University of Zagreb, Croatia; and Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands.
| | | |
Collapse
|
45
|
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: 2.4] [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]
|
46
|
Ferrer I. Sisyphus in Neverland. J Alzheimers Dis 2018; 62:1023-1047. [PMID: 29154280 PMCID: PMC5870014 DOI: 10.3233/jad-170609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
The study of life and living organisms and the way in which these interact and organize to form social communities have been central to my career. I have been fascinated by biology, neurology, and neuropathology, but also by history, sociology, and art. Certain current historical, political, and social events, some occurring proximally but others affecting people in apparently distant places, have had an impact on me. Epicurus, Seneca, and Camus shared their philosophical positions which I learned from. Many scientists from various disciplines have been exciting sources of knowledge as well. I have created a world of hypothesis and experiments but I have also got carried away by serendipity following unexpected observations. It has not been an easy path; errors and wanderings are not uncommon, and opponents close to home much more abundant than one might imagine. Ambition, imagination, resilience, and endurance have been useful in moving ahead in response to setbacks. In the end, I have enjoyed my dedication to science and I am grateful to have glimpsed beauty in it. These are brief memories of a Spanish neuropathologist born and raised in Barcelona, EU.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Service of Pathological Anatomy, Bellvitge University Hospital; CIBERNED; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
47
|
Gaitán-Peñas H, Apaja PM, Arnedo T, Castellanos A, Elorza-Vidal X, Soto D, Gasull X, Lukacs GL, Estévez R. Leukoencephalopathy-causing CLCN2 mutations are associated with impaired Cl - channel function and trafficking. J Physiol 2017; 595:6993-7008. [PMID: 28905383 DOI: 10.1113/jp275087] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/11/2017] [Indexed: 01/30/2023] Open
Abstract
KEY POINTS Characterisation of most mutations found in CLCN2 in patients with CC2L leukodystrophy show that they cause a reduction in function of the chloride channel ClC-2. GlialCAM, a regulatory subunit of ClC-2 in glial cells and involved in the leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC), increases the activity of a ClC-2 mutant by affecting ClC-2 gating and by stabilising the mutant at the plasma membrane. The stabilisation of ClC-2 at the plasma membrane by GlialCAM depends on its localisation at cell-cell junctions. The membrane protein MLC1, which is defective in MLC, also contributes to the stabilisation of ClC-2 at the plasma membrane, providing further support for the view that GlialCAM, MLC1 and ClC-2 form a protein complex in glial cells. ABSTRACT Mutations in CLCN2 have been recently identified in patients suffering from a type of leukoencephalopathy involving intramyelinic oedema. Here, we characterised most of these mutations that reduce the function of the chloride channel ClC-2 and impair its plasma membrane (PM) expression. Detailed biochemical and electrophysiological analyses of the Ala500Val mutation revealed that defective gating and increased cellular and PM turnover contributed to defective A500V-ClC-2 functional expression. Co-expression of the adhesion molecule GlialCAM, which forms a tertiary complex with ClC-2 and megalencephalic leukoencephalopathy with subcortical cysts 1 (MLC1), rescued the functional expression of the mutant by modifying its gating properties. GlialCAM also restored the PM levels of the channel by impeding its turnover at the PM. This rescue required ClC-2 localisation to cell-cell junctions, since a GlialCAM mutant with compromised junctional localisation failed to rescue the impaired stability of mutant ClC-2 at the PM. Wild-type, but not mutant, ClC-2 was also stabilised by MLC1 overexpression. We suggest that leukodystrophy-causing CLCN2 mutations reduce the functional expression of ClC-2, which is partly counteracted by GlialCAM/MLC1-mediated increase in the gating and stability of the channel.
Collapse
Affiliation(s)
- Héctor Gaitán-Peñas
- 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, Spain
| | - Pirjo M Apaja
- Department of Physiology, McGill University, Montréal, Quebec, H3E 1Y6, Canada.,Research Group Focused on Protein Structure, McGill University, Montréal, Quebec, H3E 1Y6, Canada.,South Australian Health and Medical Research Institute, Nutrition and Metabolism Theme and EMBL Australia, 5000, Adelaide, Australia
| | - Tanit Arnedo
- 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, Spain
| | - Aida Castellanos
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School, Institute of Neurosciences, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - 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, Spain
| | - David Soto
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School, Institute of Neurosciences, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School, Institute of Neurosciences, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Montréal, Quebec, H3E 1Y6, Canada.,Research Group Focused on Protein Structure, McGill University, Montréal, Quebec, H3E 1Y6, Canada
| | - Raúl Estévez
- 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, Spain
| |
Collapse
|
48
|
Ferrer I. Diversity of astroglial responses across human neurodegenerative disorders and brain aging. Brain Pathol 2017; 27:645-674. [PMID: 28804999 PMCID: PMC8029391 DOI: 10.1111/bpa.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Astrogliopathy refers to alterations of astrocytes occurring in diseases of the nervous system, and it implies the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Reactive astrocytosis refers to the response of astrocytes to different insults to the nervous system, whereas astrocytopathy indicates hypertrophy, atrophy/degeneration and loss of function and pathological remodeling occurring as a primary cause of a disease or as a factor contributing to the development and progression of a particular disease. Reactive astrocytosis secondary to neuron loss and astrocytopathy due to intrinsic alterations of astrocytes occur in neurodegenerative diseases, overlap each other, and, together with astrocyte senescence, contribute to disease-specific astrogliopathy in aging and neurodegenerative diseases with abnormal protein aggregates in old age. In addition to the well-known increase in glial fibrillary acidic protein and other proteins in reactive astrocytes, astrocytopathy is evidenced by deposition of abnormal proteins such as β-amyloid, hyper-phosphorylated tau, abnormal α-synuclein, mutated huntingtin, phosphorylated TDP-43 and mutated SOD1, and PrPres , in Alzheimer's disease, tauopathies, Lewy body diseases, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease, respectively. Astrocytopathy in these diseases can also be manifested by impaired glutamate transport; abnormal metabolism and release of neurotransmitters; altered potassium, calcium and water channels resulting in abnormal ion and water homeostasis; abnormal glucose metabolism; abnormal lipid and, particularly, cholesterol metabolism; increased oxidative damage and altered oxidative stress responses; increased production of cytokines and mediators of the inflammatory response; altered expression of connexins with deterioration of cell-to-cell networks and transfer of gliotransmitters; and worsening function of the blood brain barrier, among others. Increased knowledge of these aspects will permit a better understanding of brain aging and neurodegenerative diseases in old age as complex disorders in which neurons are not the only players.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- Institute of NeuropathologyPathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos IIIMadridSpain
| |
Collapse
|
49
|
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: 252] [Impact Index Per Article: 31.5] [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
|
50
|
Choi SA, Kim SY, Yoon J, Choi J, Park SS, Seong MW, Kim H, Hwang H, Choi JE, Chae JH, Kim KJ, Kim S, Lee YJ, Nam SO, Lim BC. A Unique Mutational Spectrum of MLC1 in Korean Patients With Megalencephalic Leukoencephalopathy With Subcortical Cysts: p.Ala275Asp Founder Mutation and Maternal Uniparental Disomy of Chromosome 22. Ann Lab Med 2017; 37:516-521. [PMID: 28840990 PMCID: PMC5587825 DOI: 10.3343/alm.2017.37.6.516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/18/2017] [Accepted: 06/25/2017] [Indexed: 11/30/2022] Open
Abstract
Background Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare inherited disorder characterized by infantile-onset macrocephaly, slow neurologic deterioration, and seizures. Mutations in the causative gene, MLC1, are found in approximately 75% of patients and are inherited in an autosomal recessive manner. We analyzed MLC1 mutations in five unrelated Korean patients with MLC. Methods Direct Sanger sequencing was used to identify MLC1 mutations. A founder effect of the p.Ala275Asp variant was demonstrated by haplotype analysis using single-nucleotide polymorphic (SNP) markers. Multiple ligation-dependent probe amplification (MLPA) and comparative genomic hybridization plus SNP array were used to detect exonic deletions or uniparental disomy (UPD). Results The most prevalent pathogenic variant was c.824C>A (p.Ala275Asp) found in 7/10 (70%) alleles. Two pathogenic frameshift variants were found: c.135delC (p.Cys46Alafs*12) and c.337_353delinsG (p.Ile113Glyfs*4). Haplotype analysis suggested that the Korean patients with MLC harbored a founder mutation in p.Ala275Asp. The p.(Ile113Glyfs*4) was identified in a homozygous state, and a family study revealed that only the mother was heterozygous for this variant. Further analysis of MLPA and SNP arrays for this patient demonstrated loss of heterozygosity of chromosome 22 without any deletion, indicating UPD. The maternal origin of both chromosomes 22 was demonstrated by haplotype analysis. Conclusions This study is the first to describe the mutational spectrum of Korean patients with MLC, demonstrating a founder effect of the p.Ala275Asp variant. This study also broadens our understanding of the mutational spectrum of MLC1 by demonstrating a homozygous p.(Ile113Glyfs*4) variant resulting from UPD of chromosome 22.
Collapse
Affiliation(s)
- Sun Ah Choi
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jihoo Yoon
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Joongmoon Choi
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Sup Park
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Moon Woo Seong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.
| | - Hunmin Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hee Hwang
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Eun Choi
- Department of Pediatrics, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jong Hee Chae
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Joong Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seunghyo Kim
- Department of Pediatrics, Jeju National University Hospital, Jeju National University School of Medicine, Jeju, Korea
| | - Yun Jin Lee
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University College of Medicine, Yangsan, Korea
| | - Sang Ook Nam
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University College of Medicine, Yangsan, Korea
| | - Byung Chan Lim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|