1
|
Christou M, Sargiannidou I, Papacharalambous R, Richter J, Tryfonos C, Christodoulou C, Kagiava A, Kleopa KA. A dose escalation and safety study of AAVrh10-mediated Schwann cell-targeted gene therapy for CMT1X. Neurotherapeutics 2025; 22:e00568. [PMID: 40055046 DOI: 10.1016/j.neurot.2025.e00568] [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: 09/08/2024] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025] Open
Abstract
X-linked Charcot-Marie-Tooth disease (CMT1X) is an inherited demyelinating neuropathy caused by loss-of-function mutations in the GJB1 gene, encoding the gap junction protein connexin32 (Cx32). Cx32 plays a critical role in Schwann cell function and myelin formation in the peripheral nervous system. We have developed a gene replacement therapeutic approach using a humanized AAVrh10 vector construct expressing GJB1 under the control of the Schwann cell-specific human myelin protein zero (MPZ) promoter. Lumbar intrathecal injection of increasing AAVrh10-hMPZ.GJB1 doses (low: 1 × 1011 vg, standard: 2 × 1011 vg and high: 1 × 1012 vg) into Gjb1-null mice resulted in adequate, dose-dependent biodistribution of the vector in anterior lumbar roots and peripheral nerves, as well as high rates of Schwann cell-specific Cx32 expression in the standard- and high-dose groups. Both standard and high vector doses provided significant therapeutic benefit evaluated by behavioural, electrophysiological and morphological outcomes. Intrathecal delivery of AAVrh10-hMPZ.GJB1 induced the production of anti-AAVrh10 antibodies at 6 weeks post-injection. However, no histopathological or inflammatory changes were observed in neural or peripheral tissues, besides a mild increase in inflammatory cell numbers in sciatic nerves of mice treated with the high dose only. This study provides proof of concept for a clinically translatable AAVrh10-mediated gene therapy approach for CMT1X.
Collapse
Affiliation(s)
- Melina Christou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Revekka Papacharalambous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus; Centre for Neuromuscular Disorders and Neuropathology Lab, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Jan Richter
- Molecular Virology Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Christina Tryfonos
- Molecular Virology Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Christina Christodoulou
- Molecular Virology Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| | - Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus.
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus; Centre for Neuromuscular Disorders and Neuropathology Lab, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Nicosia, Cyprus
| |
Collapse
|
2
|
Epel B, Viswakarma N, Hameed S, Freidin MM, Abrams CK, Kotecha M. Assessment of blood-brain barrier leakage and brain oxygenation in Connexin-32 knockout mice with systemic neuroinflammation using pulse electron paramagnetic resonance imaging techniques. Magn Reson Med 2024; 91:2519-2531. [PMID: 38193348 PMCID: PMC10997480 DOI: 10.1002/mrm.29994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE The determination of blood-brain barrier (BBB) integrity and partial pressure of oxygen (pO2) in the brain is of substantial interest in several neurological applications. This study aimed to assess the feasibility of using trityl OX071-based pulse electron paramagnetic resonance imaging (pEPRI) to provide a quantitative estimate of BBB integrity and pO2 maps in mouse brains as a function of neuroinflammatory disease progression. METHODS Five Connexin-32 (Cx32)-knockout (KO) mice were injected with lipopolysaccharide to induce neuroinflammation for imaging. Three wild-type mice were also used to optimize the imaging procedure and as control animals. An additional seven Cx32-KO mice were used to establish the BBB leakage of trityl using the colorimetric assay. All pEPRI experiments were performed using a preclinical instrument, JIVA-25 (25 mT/720 MHz), at times t = 0, 4, and 6 h following lipopolysaccharide injection. Two pEPRI imaging techniques were used: (a) single-point imaging for obtaining spatial maps to outline the brain and calculate BBB leakage using the signal amplitude, and (b) inversion-recovery electron spin echo for obtaining pO2 maps. RESULTS A statistically significant change in BBB leakage was found using pEPRI with the progression of inflammation in Cx32 KO animals. However, the change in pO2 values with the progression of inflammation for these animals was not statistically significant. CONCLUSIONS For the first time, we show the ability of pEPRI to provide pO2 maps in mouse brains noninvasively, along with a quantitative assessment of BBB leakage. We expect this study to open new queries from the field to explore the pathology of many neurological diseases and provide a path to new treatments.
Collapse
Affiliation(s)
- Boris Epel
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Illinois, USA
| | - Navin Viswakarma
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
| | - Safa Hameed
- Oxygen Measurement Core, O2M Technologies, LLC, Chicago, Illinois, USA
| | - Mona M Freidin
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Charles K Abrams
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois, USA
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
3
|
Zhan F, Tian W, Cao Y, Wu J, Ni R, Liu T, Yuan Y, Luan X, Cao L. Episodic Neurological Dysfunction in X-Linked Charcot-Marie-Tooth Disease: Expansion of the Phenotypic and Genetic Spectrum. J Clin Neurol 2024; 20:59-66. [PMID: 38179633 PMCID: PMC10782082 DOI: 10.3988/jcn.2023.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND AND PURPOSE X-linked Charcot-Marie-Tooth disease type 1 (CMTX1) is characterized by peripheral neuropathy with or without episodic neurological dysfunction. We performed clinical, neuropathological, and genetic investigations of a series of patients with mutations of the gap-junction beta-1 gene (GJB1) to extend the phenotypic and genetic description of CMTX1. METHODS Detailed clinical evaluations, sural nerve biopsy, and genetic analysis were applied to patients with CMTX1. RESULTS We collected 27 patients with CMTX1 with GJB1 mutations from 14 unrelated families. The age at onset (AAO) was 20.9±12.2 years (mean±standard deviation; range, 2-45 years). Walking difficulties, weakness in the legs, and pes cavus were common initial symptoms. Compared with female patients, males tended to have a younger AAO (males vs. females=15.4±9.6 vs. 32.0±8.8 years, p=0.002), a longer disease course (16.8±16.1 vs. 5.5±3.8 years, p=0.034), and more-severe electrophysiological results. Besides peripheral neuropathy, six of the patients had special episodic central nervous system (CNS) evidence from symptoms, signs, and/or reversible white-matter lesions. Neuropathology revealed the loss of large myelinated fibers, increased number of regenerated axon clusters with abnormally thin myelin sheaths, and excessively folded myelin. Genetic analysis identified 14 GJB1 variants, 6 of which were novel. CONCLUSIONS These findings expand the phenotypic and genetic spectrum of CMTX1. Although CMTX1 was found to have high phenotypic and CNS involvement variabilities, detailed neurological examinations and nerve conduction studies will provide critical clues for accurate diagnoses. Further exploration of the underlying mechanisms of connexin 32 involvement in neuropathy or CNS dysfunction is warranted to develop promising therapies.
Collapse
Affiliation(s)
- Feixia Zhan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wotu Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwen Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingying Wu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilong Ni
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Taotao Liu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xinghua Luan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Li Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China.
| |
Collapse
|
4
|
Kagiava A, Karaiskos C, Lapathitis G, Heslegrave A, Sargiannidou I, Zetterberg H, Bosch A, Kleopa KA. Gene replacement therapy in two Golgi-retained CMT1X mutants before and after the onset of demyelinating neuropathy. Mol Ther Methods Clin Dev 2023; 30:377-393. [PMID: 37645436 PMCID: PMC10460951 DOI: 10.1016/j.omtm.2023.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
X-linked Charcot-Marie-Tooth disease type 1 (CMT1X) is a demyelinating neuropathy resulting from loss-of-function mutations affecting the GJB1/connexin 32 (Cx32) gene. We previously showed functional and morphological improvement in Gjb1-null mice following AAV9-mediated delivery of human Cx32 driven by the myelin protein zero (Mpz) promoter in Schwann cells. However, CMT1X mutants may interfere with virally delivered wild-type (WT) Cx32. To confirm the efficacy of this vector also in the presence of CMT1X mutants, we delivered AAV9-Mpz-GJB1 by lumbar intrathecal injection in R75W/Gjb1-null and N175D/Gjb1-null transgenic lines expressing Golgi-retained mutations, before and after the onset of the neuropathy. Widespread expression of virally delivered Cx32 was demonstrated in both genotypes. Re-establishment of WT Cx32 function resulted in improved muscle strength and increased sciatic nerve motor conduction velocities in all treated groups from both mutant lines when treated before as well as after the onset of the neuropathy. Furthermore, morphological analysis showed improvement of myelination and reduction of inflammation in lumbar motor roots and peripheral nerves. In conclusion, this study provides proof of principle for a clinically translatable gene therapy approach to treat CMT1X before and after the onset of the neuropathy, even in the presence of endogenously expressed Golgi-retained Cx32 mutants.
Collapse
Affiliation(s)
- Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Christos Karaiskos
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - George Lapathitis
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, 40530 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 40530 Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Assumpció Bosch
- Department of Biochemistry & Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Bellatera, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 028029 Madrid, Spain
| | - Kleopas A. Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
- Center for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| |
Collapse
|
5
|
Wu C, Wang M, Wang X, Li W, Li S, Chen B, Niu S, Tai H, Pan H, Zhang Z. The genetic and phenotypic spectra of adult genetic leukoencephalopathies in a cohort of 309 patients. Brain 2023; 146:2364-2376. [PMID: 36380532 PMCID: PMC10232248 DOI: 10.1093/brain/awac426] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/30/2022] [Accepted: 11/01/2022] [Indexed: 08/12/2023] Open
Abstract
Genetic leukoencephalopathies (gLEs) are a highly heterogeneous group of rare genetic disorders. The spectrum of gLEs varies among patients of different ages. Distinct from the relatively more abundant studies of gLEs in children, only a few studies that explore the spectrum of adult gLEs have been published, and it should be noted that the majority of these excluded certain gLEs. Thus, to date, no large study has been designed and conducted to characterize the genetic and phenotypic spectra of gLEs in adult patients. We recruited a consecutive series of 309 adult patients clinically suspected of gLEs from Beijing Tiantan Hospital between January 2014 and December 2021. Whole-exome sequencing, mitochondrial DNA sequencing and repeat analysis of NOTCH2NLC, FMR1, DMPK and ZNF9 were performed for patients. We describe the genetic and phenotypic spectra of the set of patients with a genetically confirmed diagnosis and summarize their clinical and radiological characteristics. A total of 201 patients (65%) were genetically diagnosed, while 108 patients (35%) remained undiagnosed. The most frequent diseases were leukoencephalopathies related to NOTCH3 (25%), NOTCH2NLC (19%), ABCD1 (9%), CSF1R (7%) and HTRA1 (5%). Based on a previously proposed pathological classification, the gLEs in our cohort were divided into leukovasculopathies (35%), leuko-axonopathies (31%), myelin disorders (21%), microgliopathies (7%) and astrocytopathies (6%). Patients with NOTCH3 mutations accounted for 70% of the leukovasculopathies, followed by HTRA1 (13%) and COL4A1/2 (9%). The leuko-axonopathies contained the richest variety of associated genes, of which NOTCH2NLC comprised 62%. Among myelin disorders, demyelinating leukoencephalopathies (61%)-mainly adrenoleukodystrophy and Krabbe disease-accounted for the majority, while hypomyelinating leukoencephalopathies (2%) were rare. CSF1R was the only mutated gene detected in microgliopathy patients. Leukoencephalopathy with vanishing white matter disease due to mutations in EIF2B2-5 accounted for half of the astrocytopathies. We characterized the genetic and phenotypic spectra of adult gLEs in a large Chinese cohort. The most frequently mutated genes were NOTCH3, NOTCH2NLC, ABCD1, CSF1R and HTRA1.
Collapse
Affiliation(s)
- Chujun Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Mengwen Wang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 350005 Fuzhou, China
| | - Xingao Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Wei Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Shaowu Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Bin Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Songtao Niu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Hongfei Tai
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Hua Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Zaiqiang Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
- China National Clinical Research Centre for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| |
Collapse
|
6
|
Jennings MJ, Kagiava A, Vendredy L, Spaulding EL, Stavrou M, Hathazi D, Grüneboom A, De Winter V, Gess B, Schara U, Pogoryelova O, Lochmüller H, Borchers CH, Roos A, Burgess RW, Timmerman V, Kleopa KA, Horvath R. NCAM1 and GDF15 are biomarkers of Charcot-Marie-Tooth disease in patients and mice. Brain 2022; 145:3999-4015. [PMID: 35148379 PMCID: PMC9679171 DOI: 10.1093/brain/awac055] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/22/2021] [Accepted: 12/15/2021] [Indexed: 02/02/2023] Open
Abstract
Molecular markers scalable for clinical use are critical for the development of effective treatments and the design of clinical trials. Here, we identify proteins in sera of patients and mouse models with Charcot-Marie-Tooth disease (CMT) with characteristics that make them suitable as biomarkers in clinical practice and therapeutic trials. We collected serum from mouse models of CMT1A (C61 het), CMT2D (GarsC201R, GarsP278KY), CMT1X (Gjb1-null), CMT2L (Hspb8K141N) and from CMT patients with genotypes including CMT1A (PMP22d), CMT2D (GARS), CMT2N (AARS) and other rare genetic forms of CMT. The severity of neuropathy in the patients was assessed by the CMT Neuropathy Examination Score (CMTES). We performed multitargeted proteomics on both sample sets to identify proteins elevated across multiple mouse models and CMT patients. Selected proteins and additional potential biomarkers, such as growth differentiation factor 15 (GDF15) and cell free mitochondrial DNA, were validated by ELISA and quantitative PCR, respectively. We propose that neural cell adhesion molecule 1 (NCAM1) is a candidate biomarker for CMT, as it was elevated in Gjb1-null, Hspb8K141N, GarsC201R and GarsP278KY mice as well as in patients with both demyelinating (CMT1A) and axonal (CMT2D, CMT2N) forms of CMT. We show that NCAM1 may reflect disease severity, demonstrated by a progressive increase in mouse models with time and a significant positive correlation with CMTES neuropathy severity in patients. The increase in NCAM1 may reflect muscle regeneration triggered by denervation, which could potentially track disease progression or the effect of treatments. We found that member proteins of the complement system were elevated in Gjb1-null and Hspb8K141N mouse models as well as in patients with both demyelinating and axonal CMT, indicating possible complement activation at the impaired nerve terminals. However, complement proteins did not correlate with the severity of neuropathy measured on the CMTES scale. Although the complement system does not seem to be a prognostic biomarker, we do show complement elevation to be a common disease feature of CMT, which may be of interest as a therapeutic target. We also identify serum GDF15 as a highly sensitive diagnostic biomarker, which was elevated in all CMT genotypes as well as in Hspb8K141N, Gjb1-null, GarsC201R and GarsP278KY mouse models. Although we cannot fully explain its origin, it may reflect increased stress response or metabolic disturbances in CMT. Further large and longitudinal patient studies should be performed to establish the value of these proteins as diagnostic and prognostic molecular biomarkers for CMT.
Collapse
Affiliation(s)
- Matthew J Jennings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Alexia Kagiava
- Department of Neuroscience and Neuromuscular Disorders Centre, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Leen Vendredy
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge, University of Antwerp, Antwerp, Belgium
| | - Emily L Spaulding
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Marina Stavrou
- Department of Neuroscience and Neuromuscular Disorders Centre, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Denisa Hathazi
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Dortmund, Germany
| | - Vicky De Winter
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge, University of Antwerp, Antwerp, Belgium
| | - Burkhard Gess
- Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Ulrike Schara
- Centre for Neuromuscular Disorders in Children, University of Duisburg-Essen, Essen, Germany
| | - Oksana Pogoryelova
- Directorate of Neurosciences, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals, NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Hanns Lochmüller
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Brain and Mind Research Institute and Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center–University of Freiburg, Faculty of Medicine, Freiburg, Germany
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Andreas Roos
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Brain and Mind Research Institute and Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Robert W Burgess
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge, University of Antwerp, Antwerp, Belgium
| | - Kleopas A Kleopa
- Department of Neuroscience and Neuromuscular Disorders Centre, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Kagiava A, Karaiskos C, Richter J, Tryfonos C, Jennings MJ, Heslegrave AJ, Sargiannidou I, Stavrou M, Zetterberg H, Reilly MM, Christodoulou C, Horvath R, Kleopa KA. AAV9-mediated Schwann cell-targeted gene therapy rescues a model of demyelinating neuropathy. Gene Ther 2021; 28:659-675. [PMID: 33692503 PMCID: PMC8599011 DOI: 10.1038/s41434-021-00250-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 01/31/2023]
Abstract
Mutations in the GJB1 gene, encoding the gap junction (GJ) protein connexin32 (Cx32), cause X-linked Charcot-Marie-Tooth disease (CMT1X), an inherited demyelinating neuropathy. We developed a gene therapy approach for CMT1X using an AAV9 vector to deliver the GJB1/Cx32 gene under the myelin protein zero (Mpz) promoter for targeted expression in Schwann cells. Lumbar intrathecal injection of the AAV9-Mpz.GJB1 resulted in widespread biodistribution in the peripheral nervous system including lumbar roots, sciatic and femoral nerves, as well as in Cx32 expression in the paranodal non-compact myelin areas of myelinated fibers. A pre-, as well as post-onset treatment trial in Gjb1-null mice, demonstrated improved motor performance and sciatic nerve conduction velocities along with improved myelination and reduced inflammation in peripheral nerve tissues. Blood biomarker levels were also significantly ameliorated in treated mice. This study provides evidence that a clinically translatable AAV9-mediated gene therapy approach targeting Schwann cells could potentially treat CMT1X.
Collapse
Affiliation(s)
- Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christos Karaiskos
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Jan Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christina Tryfonos
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Matthew J Jennings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Amanda J Heslegrave
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mary M Reilly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Christina Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus.
- Center for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus.
| |
Collapse
|
8
|
Rawat A, Morrison BM. Metabolic Transporters in the Peripheral Nerve-What, Where, and Why? Neurotherapeutics 2021; 18:2185-2199. [PMID: 34773210 PMCID: PMC8804006 DOI: 10.1007/s13311-021-01150-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
Cellular metabolism is critical not only for cell survival, but also for cell fate, function, and intercellular communication. There are several different metabolic transporters expressed in the peripheral nervous system, and they each play important roles in maintaining cellular energy. The major source of energy in the peripheral nervous system is glucose, and glucose transporters 1 and 3 are expressed and allow blood glucose to be imported and utilized by peripheral nerves. There is also increasing evidence that other sources of energy, particularly monocarboxylates such as lactate that are transported primarily by monocarboxylate transporters 1 and 2 in peripheral nerves, can be efficiently utilized by peripheral nerves. Finally, emerging evidence supports an important role for connexins and possibly pannexins in the supply and regulation of metabolic energy. In this review, we will first define these critical metabolic transporter subtypes and then examine their localization in the peripheral nervous system. We will subsequently discuss the evidence, which comes both from experiments in animal models and observations from human diseases, supporting critical roles played by these metabolic transporters in the peripheral nervous system. Despite progress made in understanding the function of these transporters, many questions and some discrepancies remain, and these will also be addressed throughout this review. Peripheral nerve metabolism is fundamentally important and renewed interest in these pathways should help to answer many of these questions and potentially provide new treatments for neurologic diseases that are partly, or completely, caused by disruption of metabolism.
Collapse
Affiliation(s)
- Atul Rawat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brett M Morrison
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Stavrou M, Sargiannidou I, Georgiou E, Kagiava A, Kleopa KA. Emerging Therapies for Charcot-Marie-Tooth Inherited Neuropathies. Int J Mol Sci 2021; 22:6048. [PMID: 34205075 PMCID: PMC8199910 DOI: 10.3390/ijms22116048] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited neuropathies known as Charcot-Marie-Tooth (CMT) disease are genetically heterogeneous disorders affecting the peripheral nerves, causing significant and slowly progressive disability over the lifespan. The discovery of their diverse molecular genetic mechanisms over the past three decades has provided the basis for developing a wide range of therapeutics, leading to an exciting era of finding treatments for this, until now, incurable group of diseases. Many treatment approaches, including gene silencing and gene replacement therapies, as well as small molecule treatments are currently in preclinical testing while several have also reached clinical trial stage. Some of the treatment approaches are disease-specific targeted to the unique disease mechanism of each CMT form, while other therapeutics target common pathways shared by several or all CMT types. As promising treatments reach the stage of clinical translation, optimal outcome measures, novel biomarkers and appropriate trial designs are crucial in order to facilitate successful testing and validation of novel treatments for CMT patients.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Elena Georgiou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Kleopas A. Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
- Center for Neuromuscular Diseases, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| |
Collapse
|
11
|
Boutary S, Echaniz-Laguna A, Adams D, Loisel-Duwattez J, Schumacher M, Massaad C, Massaad-Massade L. Treating PMP22 gene duplication-related Charcot-Marie-Tooth disease: the past, the present and the future. Transl Res 2021; 227:100-111. [PMID: 32693030 DOI: 10.1016/j.trsl.2020.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 12/30/2022]
Abstract
Charcot-Marie-Tooth (CMT) disease is the most frequent inherited neuropathy, affecting 1/1500 to 1/10000. CMT1A represents 60%-70% of all CMT and is caused by a duplication on chromosome 17p11.2 leading to an overexpression of the Peripheral Myelin Protein 22 (PMP22). PMP22 gene is under tight regulation and small changes in its expression influences myelination and affect motor and sensory functions. To date, CMT1A treatment is symptomatic and classic pharmacological options have been disappointing. Here, we review the past, present, and future treatment options for CMT1A, with a special emphasis on the highly promising potential of PMP22-targeted small interfering RNA and antisense oligonucleotides.
Collapse
Affiliation(s)
- Suzan Boutary
- U 1195, INSERM and Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Andoni Echaniz-Laguna
- U 1195, INSERM and Paris-Saclay University, Le Kremlin-Bicêtre, France; Neurology Department, AP-HP, Paris-Saclay Universityand French Referent Center for Familial Amyloid Polyneuropathy and Other Rare Peripheral Neuropathies (CRMR-NNERF), Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - David Adams
- U 1195, INSERM and Paris-Saclay University, Le Kremlin-Bicêtre, France; Neurology Department, AP-HP, Paris-Saclay Universityand French Referent Center for Familial Amyloid Polyneuropathy and Other Rare Peripheral Neuropathies (CRMR-NNERF), Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Julien Loisel-Duwattez
- U 1195, INSERM and Paris-Saclay University, Le Kremlin-Bicêtre, France; Neurology Department, AP-HP, Paris-Saclay Universityand French Referent Center for Familial Amyloid Polyneuropathy and Other Rare Peripheral Neuropathies (CRMR-NNERF), Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | | | - Charbel Massaad
- Faculty of Basic and Biomedical Sciences, Paris Descartes University, INSERM UMRS 1124, Paris, France
| | | |
Collapse
|
12
|
Mozafari S, Deboux C, Laterza C, Ehrlich M, Kuhlmann T, Martino G, Baron-Van Evercooren A. Beneficial contribution of induced pluripotent stem cell-progeny to Connexin 47 dynamics during demyelination-remyelination. Glia 2020; 69:1094-1109. [PMID: 33301181 PMCID: PMC7984339 DOI: 10.1002/glia.23950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Oligodendrocytes are extensively coupled to astrocytes, a phenomenon ensuring glial homeostasis and maintenance of central nervous system myelin. Molecular disruption of this communication occurs in demyelinating diseases such as multiple sclerosis. Less is known about the vulnerability and reconstruction of the panglial network during adult demyelination‐remyelination. Here, we took advantage of lysolcithin‐induced demyelination to investigate the expression dynamics of the oligodendrocyte specific connexin 47 (Cx47) and to some extent that of astrocyte Cx43, and whether this dynamic could be modulated by grafted induced pluripotent stem cell (iPSC)‐neural progeny. Our data show that disruption of Cx43‐Cx47 mediated hetero‐cellular gap‐junction intercellular communication following demyelination is larger in size than demyelination. Loss of Cx47 expression is timely rescued during remyelination and accelerated by the grafted neural precursors. Moreover, mouse and human iPSC‐derived oligodendrocytes express Cx47, which co‐labels with astrocyte Cx43, indicating their integration into the panglial network. These data suggest that in rodents, full lesion repair following transplantation occurs by panglial reconstruction in addition to remyelination. Targeting panglial elements by cell therapy or pharmacological compounds may help accelerating or stabilizing re/myelination in myelin disorders.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cyrille Deboux
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cecilia Laterza
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy.,Industrial Engineering Department, University of Padova, Padova, Italy
| | - Marc Ehrlich
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Anne Baron-Van Evercooren
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
13
|
Mozafari S, Starost L, Manot-Saillet B, Garcia-Diaz B, Xu YKT, Roussel D, Levy MJF, Ottoboni L, Kim KP, Schöler HR, Kennedy TE, Antel JP, Martino G, Angulo MC, Kuhlmann T, Baron-Van Evercooren A. Multiple sclerosis iPS-derived oligodendroglia conserve their properties to functionally interact with axons and glia in vivo. SCIENCE ADVANCES 2020; 6:6/49/eabc6983. [PMID: 33277253 PMCID: PMC7821889 DOI: 10.1126/sciadv.abc6983] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 05/04/2023]
Abstract
Remyelination failure in multiple sclerosis (MS) is associated with a migration/differentiation block of oligodendroglia. The reason for this block is highly debated. It could result from disease-related extrinsic or intrinsic regulators in oligodendroglial biology. To avoid confounding immune-mediated extrinsic effect, we used an immune-deficient mouse model to compare induced pluripotent stem cell-derived oligodendroglia from MS and healthy donors following engraftment in the developing CNS. We show that the MS-progeny behaves and differentiates into oligodendrocytes to the same extent as controls. They generate equal amounts of myelin, with bona fide nodes of Ranvier, and promote equal restoration of their host slow conduction. MS-progeny expressed oligodendrocyte- and astrocyte-specific connexins and established functional connections with donor and host glia. Thus, MS oligodendroglia, regardless of major immune manipulators, are intrinsically capable of myelination and making functional axo-glia/glia-glia connections, reinforcing the view that the MS oligodendrocyte differentiation block is not from major intrinsic oligodendroglial deficits.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Laura Starost
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Blandine Manot-Saillet
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Beatriz Garcia-Diaz
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Yu Kang T Xu
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Delphine Roussel
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Marion J F Levy
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Linda Ottoboni
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Timothy E Kennedy
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Maria Cecilia Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Anne Baron-Van Evercooren
- INSERM, U1127, F-75013 Paris, France.
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| |
Collapse
|
14
|
Genetic mechanisms of peripheral nerve disease. Neurosci Lett 2020; 742:135357. [PMID: 33249104 DOI: 10.1016/j.neulet.2020.135357] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022]
Abstract
Peripheral neuropathies of genetic etiology are a very diverse group of disorders manifesting either as non-syndromic inherited neuropathies without significant manifestations outside the peripheral nervous system, or as part of a systemic or syndromic genetic disorder. The former and most frequent group is collectively known as Charcot-Marie-Tooth disease (CMT), with prevalence as high as 1:2,500 world-wide, and has proven to be genetically highly heterogeneous. More than 100 different genes have been identified so far to cause various CMT forms, following all possible inheritance patterns. CMT causative genes belong to several common functional pathways that are essential for the integrity of the peripheral nerve. Their discovery has provided insights into the normal biology of axons and myelinating cells, and has highlighted the molecular mechanisms including both loss of function and gain of function effects, leading to peripheral nerve degeneration. Demyelinating neuropathies result from dysfunction of genes primarily affecting myelinating Schwann cells, while axonal neuropathies are caused by genes affecting mostly neurons and their long axons. Furthermore, mutation in genes expressed outside the nervous system, as in the case of inherited amyloid neuropathies, may cause peripheral neuropathy resulting from accumulation of β-structured amyloid fibrils in peripheral nerves in addition to various organs. Increasing insights into the molecular-genetic mechanisms have revealed potential therapeutic targets. These will enable the development of novel therapeutics for genetic neuropathies that remain, in their majority, without effective treatment.
Collapse
|
15
|
Papaneophytou C, Georgiou E, Kleopa KA. The role of oligodendrocyte gap junctions in neuroinflammation. Channels (Austin) 2020; 13:247-263. [PMID: 31232168 PMCID: PMC6602578 DOI: 10.1080/19336950.2019.1631107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Gap junctions (GJs) provide channels for direct cell-to-cell connectivity serving the homeostasis in several organs of vertebrates including the central (CNS) and peripheral (PNS) nervous systems. GJs are composed of connexins (Cx), which show a highly distinct cellular and subcellular expression pattern. Oligodendrocytes, the myelinating cells of the CNS, are characterized by extensive GJ connectivity with each other as well as with astrocytes. The main oligodendrocyte connexins forming these GJ channels are Cx47 and Cx32. The importance of these channels has been highlighted by the discovery of human diseases caused by mutations in oligodendrocyte connexins, manifesting with leukodystrophy or transient encephalopathy. Experimental models have provided further evidence that oligodendrocyte GJs are essential for CNS myelination and homeostasis, while a strong inflammatory component has been recognized in the absence of oligodendrocyte connexins. Further studies revealed that connexins are also disrupted in multiple sclerosis (MS) brain, and in experimental models of induced inflammatory demyelination. Moreover, induced demyelination was more severe and associated with higher degree of CNS inflammation in models with oligodendrocyte GJ deficiency, suggesting that disrupted connexin expression in oligodendrocytes is not only a consequence but can also drive a pro-inflammatory environment in acquired demyelinating disorders such as MS. In this review, we summarize the current insights from human disorders as well as from genetic and acquired models of demyelination related to oligodendrocyte connexins, with the remaining challenges and perspectives.
Collapse
Affiliation(s)
- Christos Papaneophytou
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus.,b Department of Life and Health Sciences, School of Sciences and Engineering , University of Nicosia , Nicosia , Cyprus
| | - Elena Georgiou
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus
| | - Kleopas A Kleopa
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus.,c Neurology Clinics , the Cyprus Institute of Neurology and Genetics, and the Cyprus School of Molecular Medicine , Nicosia , Cyprus
| |
Collapse
|
16
|
Martin EA, Lasseigne AM, Miller AC. Understanding the Molecular and Cell Biological Mechanisms of Electrical Synapse Formation. Front Neuroanat 2020; 14:12. [PMID: 32372919 PMCID: PMC7179694 DOI: 10.3389/fnana.2020.00012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
In this review article, we will describe the recent advances made towards understanding the molecular and cell biological mechanisms of electrical synapse formation. New evidence indicates that electrical synapses, which are gap junctions between neurons, can have complex molecular compositions including protein asymmetries across joined cells, diverse morphological arrangements, and overlooked similarities with other junctions, all of which indicate new potential roles in neurodevelopmental disease. Aquatic organisms, and in particular the vertebrate zebrafish, have proven to be excellent models for elucidating the molecular mechanisms of electrical synapse formation. Zebrafish will serve as our main exemplar throughout this review and will be compared with other model organisms. We highlight the known cell biological processes that build neuronal gap junctions and compare these with the assemblies of adherens junctions, tight junctions, non-neuronal gap junctions, and chemical synapses to explore the unknown frontiers remaining in our understanding of the critical and ubiquitous electrical synapse.
Collapse
Affiliation(s)
| | | | - Adam C. Miller
- Department of Biology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| |
Collapse
|
17
|
Michaelidou K, Tsiverdis I, Erimaki S, Papadimitriou D, Amoiridis G, Papadimitriou A, Mitsias P, Zaganas I. Whole exome sequencing establishes diagnosis of Charcot-Marie-Tooth 4J, 1C, and X1 subtypes. Mol Genet Genomic Med 2020; 8:e1141. [PMID: 32022442 PMCID: PMC7196464 DOI: 10.1002/mgg3.1141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Charcot-Marie-Tooth (CMT) hereditary polyneuropathies pose a diagnostic challenge. Our aim here is to describe CMT patients diagnosed by whole exome sequencing (WES) following years of fruitless testing. METHODS/RESULTS Three patients with polyneuropathy suspected to be genetic in origin, but not harboring PMP22 gene deletion/duplication, were offered WES. The first patient, a 66-year-old man, had been suffering from progressive weakness and atrophies in the lower and upper extremities for 20 years. Due to ambiguous electrophysiological findings, immune therapies were administered to no avail. Twelve years after PMP22 deletion/duplication testing, WES revealed two pathogenic variants in the FIG4 gene (p.Ile41Thr and p.Phe598fs, respectively), as a cause of CMT 4J. The second patient, a 19-year-old man, had been suffering from hearing and gait impairment since at least his infancy, and recently presented with weakness and dystonia of the lower extremities. In this patient, WES identified the p.Leu122Val LITAF gene variant in heterozygous state, suggesting the diagnosis of CMT 1C, several years after initial genetic analyses. The third patient, a 44-year-old man, presented with progressive weakness and atrophies of the lower and upper extremities since the age of 17 years old. In this patient, WES identified the hemizygous p.Arg164Gln pathogenic variant in the GJB1 gene, establishing the diagnosis of CMT X1, 8 years after testing for PMP22 deletion/duplication. CONCLUSION Novel diagnostic techniques, such as WES, offer the possibility to decipher the cause of CMT subtypes, ending the diagnostic Odyssey of the patients and sparing them from unnecessary and potentially harmful treatments.
Collapse
Affiliation(s)
- Kleita Michaelidou
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
| | - Ioannis Tsiverdis
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
| | - Sophia Erimaki
- Neurophysiology UnitUniversity Hospital of CreteHeraklion, CreteGreece
| | | | | | | | - Panayiotis Mitsias
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
- Neurophysiology UnitUniversity Hospital of CreteHeraklion, CreteGreece
- Department of NeurologyHenry Ford Hospital/Wayne State UniversityDetroitMIUSA
| | - Ioannis Zaganas
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
| |
Collapse
|
18
|
Sargiannidou I, Kagiava A, Kleopa KA. Gene therapy approaches targeting Schwann cells for demyelinating neuropathies. Brain Res 2020; 1728:146572. [PMID: 31790684 DOI: 10.1016/j.brainres.2019.146572] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/12/2019] [Accepted: 11/26/2019] [Indexed: 11/27/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) encompasses numerous genetically heterogeneous inherited neuropathies, which together are one of the commonest neurogenetic disorders. Axonal CMT types result from mutations in neuronally expressed genes, whereas demyelinating CMT forms mostly result from mutations in genes expressed by myelinating Schwann cells. The demyelinating forms are the most common, and may be caused by dominant mutations and gene dosage effects (as in CMT1), as well as by recessive mutations and loss of function mechanisms (as in CMT4). The discovery of causative genes and increasing insights into molecular mechanisms through the study of experimental disease models has provided the basis for the development of gene therapy approaches. For demyelinating CMT, gene silencing or gene replacement strategies need to be targeted to Schwann cells. Progress in gene replacement for two different CMT forms, including CMT1X caused by GJB1 gene mutations, and CMT4C, caused by SH3TC2 gene mutations, has been made through the use of a myelin-specific promoter to restrict expression in Schwann cells, and by lumbar intrathecal delivery of lentiviral viral vectors to achieve more widespread biodistribution in the peripheral nervous system. This review summarizes the molecular-genetic mechanisms of selected demyelinating CMT neuropathies and the progress made so far, as well as the remaining challenges in the path towards a gene therapy to treat these disorders through the use of optimal gene therapy tools including clinically translatable delivery methods and adeno-associated viral (AAV) vectors.
Collapse
Affiliation(s)
- Irene Sargiannidou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Alexia Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus; Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus.
| |
Collapse
|
19
|
Kagiava A, Richter J, Tryfonos C, Karaiskos C, Heslegrave AJ, Sargiannidou I, Rossor AM, Zetterberg H, Reilly MM, Christodoulou C, Kleopa KA. Gene replacement therapy after neuropathy onset provides therapeutic benefit in a model of CMT1X. Hum Mol Genet 2019; 28:3528-3542. [PMID: 31411673 DOI: 10.1093/hmg/ddz199] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022] Open
Abstract
X-linked Charcot-Marie-Tooth disease (CMT1X), one of the commonest forms of inherited demyelinating neuropathy, results from GJB1 gene mutations causing loss of function of the gap junction protein connexin32 (Cx32). The aim of this study was to examine whether delayed gene replacement therapy after the onset of peripheral neuropathy can provide a therapeutic benefit in the Gjb1-null/Cx32 knockout model of CMT1X. After delivery of the LV-Mpz.GJB1 lentiviral vector by a single lumbar intrathecal injection into 6-month-old Gjb1-null mice, we confirmed expression of Cx32 in lumbar roots and sciatic nerves correctly localized at the paranodal myelin areas. Gjb1-null mice treated with LV-Mpz.GJB1 compared with LV-Mpz.Egfp (mock) vector at the age of 6 months showed improved motor performance at 8 and 10 months. Furthermore, treated mice showed increased sciatic nerve conduction velocities, improvement of myelination and reduced inflammation in lumbar roots and peripheral nerves at 10 months of age, along with enhanced quadriceps muscle innervation. Plasma neurofilament light (NEFL) levels, a clinically relevant biomarker, were also ameliorated in fully treated mice. Intrathecal gene delivery after the onset of peripheral neuropathy offers a significant therapeutic benefit in this disease model, providing a proof of principle for treating patients with CMT1X at different ages.
Collapse
Affiliation(s)
- A Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - J Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - C Tryfonos
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - C Karaiskos
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - A J Heslegrave
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - I Sargiannidou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - A M Rossor
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - H Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - M M Reilly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - C Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - K A Kleopa
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| |
Collapse
|
20
|
Nueva mutación genética en un caso de enfermedad de Charcot-Marie-Tooth. Neurologia 2019; 34:546-547. [DOI: 10.1016/j.nrl.2017.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 01/08/2017] [Indexed: 11/20/2022] Open
|
21
|
Domínguez Díez F, López Alburquerque J. New mutation in a patient with Charcot-Marie-Tooth disease. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2017.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
22
|
Doğan Y, Gül Ş, Ceylan AC, Kutsal YG. A special association between Charcot-Marie-Tooth type 1A disease and relapsing remitting multiple sclerosis. Mult Scler Relat Disord 2019; 35:83-85. [PMID: 31352181 DOI: 10.1016/j.msard.2019.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/12/2019] [Accepted: 07/19/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Central nervous system involvement has been reported in different subtypes of Charcot-Marie-Tooth (CMT) diseases. The increasing number of cases with CMT and MS may provide further information about the common pathway of demyelination and MS pathogenesis. CASE PRESENTATION We report the case of a 21-year-old woman with CMT1A and MS. Bilateral rest and intention tremor, steroid associated psychotic episodes, and severe disability at an early age were unexpected aspects of this case. CONCLUSION PMP22, the target protein in CMT1A, shares partial homology with other CNS proteins. PMP22 gene might be relevant to a common pathway of the demyelinating process.
Collapse
Affiliation(s)
- Yahya Doğan
- Hacettepe University Medical School, Department of Physical and Rehabilitation Medicine, Ankara, Turkey.
| | - Şule Gül
- Hacettepe University Medical School, Department of Physical and Rehabilitation Medicine, Ankara, Turkey
| | - Ahmet Cevdet Ceylan
- Atatürk Training and Research Hospital, Department of Genetics, Yıldırım Beyazıt University, Ankara, Turkey
| | - Yeşim Gökçe Kutsal
- Hacettepe University Medical School, Department of Physical and Rehabilitation Medicine, Ankara, Turkey
| |
Collapse
|
23
|
Corvino V, Apisa P, Malesci R, Laria C, Auletta G, Franzé A. X-Linked Sensorineural Hearing Loss: A Literature Review. Curr Genomics 2018; 19:327-338. [PMID: 30065609 PMCID: PMC6030855 DOI: 10.2174/1389202919666171218163046] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/10/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023] Open
Abstract
Sensorineural hearing loss is a very diffuse pathology (about 1/1000 born) with several types of transmission. X-linked hearing loss accounts for approximately 1% - 2% of cases of non-syndromic forms, as well as for many syndromic forms. To date, six loci (DFNX1-6) and five genes (PRPS1 for DFNX1, POU3F4 for DFNX2, SMPX for DFNX4, AIFM1 for DFNX5 and COL4A6 for DFNX6) have been identified for X-linked non-syndromic hearing loss. For the syndromic forms, at least 15 genes have been identified, some of which are also implicated in non-syndromic forms. Moreover, some syndromic forms, presenting large chromosomal deletions, are associated with mental retardation too. This review presents an overview of the currently known genes related to X-linked hearing loss with the support of the most recent literature. It summarizes the genetics and clinical features of X-linked hearing loss to give information useful to realize a clear genetic counseling and an early diagnosis. It is important to get an early diagnosis of these diseases to decide the investigations to predict the evolution of the disease and the onset of any other future symptoms. This information will be clearly useful for choosing the best therapeutic strategy. In particular, regarding audiological aspects, this review highlights risks and benefits currently known in some cases for specific therapeutic intervention.
Collapse
Affiliation(s)
- Virginia Corvino
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
| | - Pasqualina Apisa
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
| | - Rita Malesci
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
| | - Carla Laria
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
| | - Gennaro Auletta
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
| | - Annamaria Franzé
- Unit of Audiology, Department of Neurosciences, Reproductives and Odontostomatologic Sciences, University of Naples “Federico II”, Naples, Italy
- CEINGE Biotecnologie Avanzate, Naples, Italy
| |
Collapse
|
24
|
Bortolozzi M. What's the Function of Connexin 32 in the Peripheral Nervous System? Front Mol Neurosci 2018; 11:227. [PMID: 30042657 PMCID: PMC6048289 DOI: 10.3389/fnmol.2018.00227] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
Connexin 32 (Cx32) is a fundamental protein in the peripheral nervous system (PNS) as its mutations cause the X-linked form of Charcot–Marie–Tooth disease (CMT1X), the second most common form of hereditary motor and sensory neuropathy and a demyelinating disease for which there is no effective therapy. Since mutations of the GJB1 gene encoding Cx32 were first reported in 1993, over 450 different mutations associated with CMT1X including missense, frameshift, deletion and non-sense ones have been identified. Despite the availability of a sizable number of studies focusing on normal and mutated Cx32 channel properties, the crucial role played by Cx32 in the PNS has not yet been elucidated, as well as the molecular pathogenesis of CMT1X. Is Cx32 fundamental during a particular phase of Schwann cell (SC) life? Are Cx32 paired (gap junction, GJ) channels in myelinated SCs important for peripheral nerve homeostasis? The attractive hypothesis that short coupling of adjacent myelin layers by Cx32 GJs is required for efficient diffusion of K+ and signaling molecules is still debated, while a growing body of evidence is supporting other possible functions of Cx32 in the PNS, mainly related to Cx32 unpaired channels (hemichannels), which could be involved in a purinergic-dependent pathway controlling myelination. Here we review the intriguing puzzle of findings about Cx32 function and dysfunction, discussing possible directions for future investigation.
Collapse
Affiliation(s)
- Mario Bortolozzi
- Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padua, Italy.,Padova Neuroscience Center (PNC), Padua, Italy
| |
Collapse
|
25
|
Kagiava A, Karaiskos C, Richter J, Tryfonos C, Lapathitis G, Sargiannidou I, Christodoulou C, Kleopa KA. Intrathecal gene therapy in mouse models expressing CMT1X mutations. Hum Mol Genet 2018; 27:1460-1473. [PMID: 29462293 DOI: 10.1093/hmg/ddy056] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/10/2018] [Indexed: 11/14/2022] Open
Abstract
Gap junction beta-1 (GJB1) gene mutations affecting the gap junction protein connexin32 (Cx32) cause the X-linked Charcot-Marie-Tooth disease (CMT1X), a common inherited neuropathy. Targeted expression of virally delivered Cx32 in Schwann cells following intrathecal injection of lentiviral vectors in the Cx32 knockout (KO) mouse model of the disease has led to morphological and functional improvement. To examine whether this approach could be effective in CMT1X patients expressing different Cx32 mutants, we treated transgenic Cx32 KO mice expressing the T55I, R75W or N175D CMT1X mutations. All three mutants were localized in the perinuclear compartment of myelinating Schwann cells consistent with retention in the ER (T55I) or Golgi (R75W, N175D) and loss of physiological expression in the non-compact myelin. Following intrathecal delivery of the GJB1 gene we detected the virally delivered wild-type (WT) Cx32 in non-compact myelin of T55I KO mice, but only rarely in N175D KO or R75W KO mice, suggesting dominant-negative effects of the R75W and N175D mutants but not of the T55I mutant on co-expressed WT Cx32. GJB1 treated T55I KO mice showed improved motor performance, lower ratios of abnormally myelinated fibers and reduction of inflammatory cells in spinal roots and peripheral nerves compared with mock-treated littermates. Either partial (N175D KO) or no (R75W KO) improvement was observed in the other two mutant lines. Thus, certain CMT1X mutants may interfere with gene addition therapy for CMT1X. Whereas gene addition can be used for non-interfering CMT1X mutations, further studies will be needed to develop treatments for patients harboring interfering mutations.
Collapse
Affiliation(s)
- A Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - C Karaiskos
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - J Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - C Tryfonos
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - G Lapathitis
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - I Sargiannidou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - C Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| | - K A Kleopa
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
- Neurology Clinics, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| |
Collapse
|
26
|
Carrer A, Leparulo A, Crispino G, Ciubotaru CD, Marin O, Zonta F, Bortolozzi M. Cx32 hemichannel opening by cytosolic Ca2+ is inhibited by the R220X mutation that causes Charcot-Marie-Tooth disease. Hum Mol Genet 2018; 27:80-94. [PMID: 29077882 DOI: 10.1093/hmg/ddx386] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/17/2017] [Indexed: 11/15/2022] Open
Abstract
Mutations of the GJB1 gene encoding connexin 32 (Cx32) cause the X-linked form of Charcot-Marie-Tooth disease (CMTX1), a demyelinating peripheral neuropathy for which there is no cure. A growing body of evidence indicates that ATP release through Cx32 hemichannels in Schwann cells could be critical for nerve myelination, but it is unknown if CMTX1 mutations alter the cytosolic Ca2+-dependent gating mechanism that controls Cx32 hemichannel opening and ATP release. The current study uncovered that loss of the C-terminus in Cx32 (R220X mutation), which causes a severe CMTX1 phenotype, inhibits hemichannel opening during a canonical IP3-mediated increase in cytosolic Ca2+ in HeLa cells. Interestingly, the gating function of R220X hemichannels was completely restored by both the intracellular and extracellular application of a peptide that mimics the Cx32 cytoplasmic loop. All-atom molecular dynamics simulations suggest that loss of the C-terminus in the mutant hemichannel triggers abnormal fluctuations of the cytoplasmic loop which are prevented by binding to the mimetic peptide. Experiments that stimulated R220X hemichannel opening by cell depolarization displayed reduced voltage sensitivity with respect to wild-type hemichannels which was explained by loss of subconductance states at the single channel level. Finally, experiments of intercellular diffusion mediated by wild-type or R220X gap junction channels revealed similar unitary permeabilities to ions, signalling molecules (cAMP) or larger solutes (Lucifer yellow). Taken together, our findings support the hypothesis that paracrine signalling alteration due to Cx32 hemichannel dysfunction underlies CMTX1 pathogenesis and suggest a candidate molecule for novel studies investigating a therapeutic approach.
Collapse
Affiliation(s)
- Andrea Carrer
- Venetian Institute of Molecular Medicine (VIMM), Padua 35129, Italy
- Department of Physics and Astronomy "G. Galilei", University of Padua, Padua 35131, Italy
| | - Alessandro Leparulo
- Venetian Institute of Molecular Medicine (VIMM), Padua 35129, Italy
- Department of Physics and Astronomy "G. Galilei", University of Padua, Padua 35131, Italy
| | - Giulia Crispino
- Venetian Institute of Molecular Medicine (VIMM), Padua 35129, Italy
- Department of Physics and Astronomy "G. Galilei", University of Padua, Padua 35131, Italy
| | | | - Oriano Marin
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Monterotondo 00015, Italy
| | - Mario Bortolozzi
- Venetian Institute of Molecular Medicine (VIMM), Padua 35129, Italy
- Department of Physics and Astronomy "G. Galilei", University of Padua, Padua 35131, Italy
- Italian National Research Council (CNR), Institute of Protein Biochemistry, Naples 80131, Italy
| |
Collapse
|
27
|
Kyriakoudi S, Sargiannidou I, Kagiava A, Olympiou M, Kleopa KA. Golgi-retained Cx32 mutants interfere with gene addition therapy for CMT1X. Hum Mol Genet 2017; 26:1622-1633. [PMID: 28334782 DOI: 10.1093/hmg/ddx064] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2023] Open
Abstract
Numerous GJB1 gene mutations cause the X-linked form of Charcot-Marie-Tooth disease (CMT1X). GJB1 encodes connexin32 (Cx32), which forms trans-myelin gap junctions in Schwann cells. Most GJB1 mutations result in loss-of-function mechanisms, supporting the concept of gene replacement therapy. However, interactions between delivered wild type and endogenously expressed mutant Cx32 may potentially occur in the setting of gene replacement therapy. In order to screen for possible interactions of several representative CMT1X mutants with wild type Cx32 that may interfere with the functional gap junction formation, we established an in vitro screening method co-expressing in HeLa cells wild type Cx32 and one of eight different Cx32 mutants including A39P, A39V, T55I, R75W, M93V, L143P, N175D and R183S. Some of the Golgi-retained mutants hindered gap junction plaque assembly by Cx32 on the cell membrane, while co-immunoprecipitation analysis revealed a partial interaction of wild type protein with Golgi-retained mutants. Dye transfer studies confirmed that Golgi-retained R75W, M93V and N175D but not endoplasmic reticulum-retained T55I had a negative effect on wild type Cx32 function. Finally, in vivo intraneural delivery of the gene encoding the wild type Cx32 in mice bearing either the T55I or R75W mutation on Cx32 knockout background showed that virally delivered protein was correctly localized in mice expressing the endoplasmic reticulum-retained T55I whereas it did not traffic normally in mice expressing the Golgi-retained R75W. Thus, certain Golgi-retained Cx32 mutants may interfere with exogenously delivered Cx32. Screening for mutant-wild type Cx32 interactions should be considered prior to planning gene addition therapy for CMT1X.
Collapse
Affiliation(s)
| | | | | | | | - Kleopas A Kleopa
- Neuroscience Laboratory
- Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 1683 Nicosia, Cyprus
| |
Collapse
|
28
|
Milley GM, Varga ET, Grosz Z, Bereznai B, Aranyi Z, Boczan J, Dioszeghy P, Kálmán B, Gal A, Molnar MJ. Three novel mutations and genetic epidemiology analysis of the Gap Junction Beta 1 (GJB1) gene among Hungarian Charcot-Marie-Tooth disease patients. Neuromuscul Disord 2016; 26:706-711. [PMID: 27544631 DOI: 10.1016/j.nmd.2016.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/14/2016] [Accepted: 07/25/2016] [Indexed: 02/03/2023]
Abstract
Pathogenic variants of the gap junction beta 1 (GJB1) gene are responsible for the Charcot-Marie-Tooth neuropathy X type 1 (CMTX1). In this study, we report the mutation frequency of GJB1 in 210 Hungarian CMT patients and the phenotype comparison between male and female CMTX1 patients. Altogether, 13 missense substitutions were found in the GJB1 gene. Among them, 10 have been previously described as pathogenic variants (p.Arg15Trp, p.Val63Ile, p.Leu89Val, p.Ala96Gly, p.Arg107Trp, p.Arg142Gln, p.Arg164Trp, p.Arg164Gln, p.Pro172Ala and p.Asn205Ser), while 3 were novel, likely pathogenic alterations (p.Val13Glu, p.Glu186Gly, p.Met194Ile). These variants were not present in controls and were predicted as disease causing by in silico analysis. The frequency of the variants was 6.7% in our cohort which refers to a common cause of hereditary neuropathy among Hungarian patients. In addition to the classical phenotype, CNS involvement was proved in 26.1% of the CMTX1 patients. GJB1 pathogenic alterations were found mainly in males but we also detected them in female probands. The statistical analysis of CMTX1 patients revealed a significant difference between the two genders regarding the age of onset, Charcot-Marie-Tooth neuropathy and examination scores.
Collapse
Affiliation(s)
- Gyorgy Mate Milley
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Edina Timea Varga
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary; Department of Neurology, University of Szeged, Szeged, Hungary
| | - Zoltan Grosz
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Benjamin Bereznai
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Aranyi
- MTA-SE NAP B Peripheral Nervous System Research Group, Dept. of Neurology, Semmelweis University, Budapest, Hungary
| | - Judit Boczan
- Department of Neurology, Medical Center, University of Debrecen, Debrecen, Hungary
| | - Peter Dioszeghy
- Department of Neurology, Andras Josa Teaching Hospital, Nyiregyhaza, Hungary
| | - Bernadette Kálmán
- University of Pecs, Faculty of Health Sciences, Pecs and Molecular Pathology, Markusovszky University Teaching Hospital, Szombathely, Hungary
| | - Aniko Gal
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
29
|
Tsai PC, Yang DM, Liao YC, Chiu TY, Kuo HC, Su YP, Guo YC, Soong BW, Lin KP, Liu YT, Lee YC. Clinical and biophysical characterization of 19 GJB1 mutations. Ann Clin Transl Neurol 2016; 3:854-865. [PMID: 27844031 PMCID: PMC5099531 DOI: 10.1002/acn3.347] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/08/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022] Open
Abstract
Objective Charcot–Marie–Tooth disease type X1 (CMTX1), which is caused by mutations in the gap junction (GJ) protein beta‐1 gene (GJB1), is the second most common form of Charcot–Marie–Tooth disease (CMT). GJB1 encodes the GJ beta‐1 protein (GJB1), which forms GJs within the myelin sheaths of peripheral nerves. The process by which GJB1 mutants cause neuropathy has not been fully elucidated. This study evaluated the biophysical characteristics of GJB1 mutants and their correlations with the clinical features of CMTX1 patients. Methods All patients with a validated GJB1 mutation were assessed using the Charcot–Marie–Tooth disease neuropathy score version 2 (CMTNS). The impacts of the mutations on the biophysical functions of GJB1 were characterized by assessing intracellular localization, expression patterns, and GJ Ca2+ permeability. Result Nineteen GJB1 mutations were identified in 24 patients with a clinical diagnosis of CMT. Six are novel mutations: p.L6S, p.I20F, p.I101Rfs*8, p.F153L, p.R215P, and p.D278V. Diverse pathological effects of the mutations were demonstrated, including reduced GJB1 expression, intracellular mislocalization, and altered GJ functions. GJB1 mutations that caused a complete loss of GJ Ca2+ permeability appeared to be associated with an earlier disease onset, whereas those resulting in preservation of GJ permeability and with predominant cell membrane expression tended to have a later onset and a milder phenotype. Interpretation This study demonstrated that the degree of loss of GJ function caused by the GJB1 mutations might contribute to the onset and severity of neuropathic symptoms in CMTX1.
Collapse
Affiliation(s)
- Pei-Chien Tsai
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan; Brain Research Center National Yang-Ming University Taipei 11221 Taiwan
| | - De-Ming Yang
- Microscopy Service Laboratory Basic Research Division Department of Medical Research and Education Taipei Veterans General Hospital Taipei 11217 Taiwan; Institute of Biophotonics School of Medical Technology & Engineering; Biophotonics and Molecular Imaging Research Center (BMIRC) National Yang-Ming University Taipei 11212 Taiwan
| | - Yi-Chu Liao
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan
| | - Tai-Yu Chiu
- Microscopy Service Laboratory Basic Research Division Department of Medical Research and Education Taipei Veterans General Hospital Taipei 11217 Taiwan; Institute of Biophotonics School of Medical Technology & Engineering; Biophotonics and Molecular Imaging Research Center (BMIRC) National Yang-Ming University Taipei 11212 Taiwan
| | - Hung-Chou Kuo
- Department of Neurology Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine Taoyuan 33302 Taiwan
| | - Yu-Ping Su
- Department of Psychiatry Cathay General Hospital Taipei 10687 Taiwan; School of Medicine Fu-Jen Catholic University Taipei 24205 Taiwan
| | - Yuh-Cherng Guo
- Institute of Clinical Medicine National Yang-Ming University Taipei 11221 Taiwan; Neuroscience Laboratory Department of Neurology China Medical University Hospital Taichung 40447 Taiwan; School of Medicine College of Medicine China Medical University Taichung 40402 Taiwan
| | - Bing-Wen Soong
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan; Brain Research Center National Yang-Ming University Taipei 11221 Taiwan
| | - Kon-Ping Lin
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan
| | - Yo-Tsen Liu
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan
| | - Yi-Chung Lee
- Department of Neurology Taipei Veterans General Hospital Taipei 11217 Taiwan; Department of Neurology National Yang-Ming University School of Medicine Taipei 11221 Taiwan; Brain Research Center National Yang-Ming University Taipei 11221 Taiwan
| |
Collapse
|
30
|
Olympiou M, Sargiannidou I, Markoullis K, Karaiskos C, Kagiava A, Kyriakoudi S, Abrams CK, Kleopa KA. Systemic inflammation disrupts oligodendrocyte gap junctions and induces ER stress in a model of CNS manifestations of X-linked Charcot-Marie-Tooth disease. Acta Neuropathol Commun 2016; 4:95. [PMID: 27585976 PMCID: PMC5009701 DOI: 10.1186/s40478-016-0369-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 11/10/2022] Open
Abstract
X-linked Charcot-Marie-Tooth disease (CMT1X) is a common form of inherited neuropathy resulting from different mutations affecting the gap junction (GJ) protein connexin32 (Cx32). A subset of CMT1X patients may additionally present with acute fulminant CNS dysfunction, typically triggered by conditions of systemic inflammation and metabolic stress. To clarify the underlying mechanisms of CNS phenotypes in CMT1X we studied a mouse model of systemic inflammation induced by lipopolysaccharide (LPS) injection to compare wild type (WT), connexin32 (Cx32) knockout (KO), and KO T55I mice expressing the T55I Cx32 mutation associated with CNS phenotypes. Following a single intraperitoneal LPS or saline (controls) injection at the age of 40-60 days systemic inflammatory response was documented by elevated TNF-α and IL-6 levels in peripheral blood and mice were evaluated 1 week after injection. Behavioral analysis showed graded impairment of motor performance in LPS treated mice, worse in KO T55I than in Cx32 KO and in Cx32 KO worse than WT. Iba1 immunostaining revealed widespread inflammation in LPS treated mice with diffusely activated microglia throughout the CNS. Immunostaining for the remaining major oligodendrocyte connexin Cx47 and for its astrocytic partner Cx43 revealed widely reduced expression of Cx43 and loss of Cx47 GJs in oligodendrocytes. Real-time PCR and immunoblot analysis indicated primarily a down regulation of Cx43 expression with secondary loss of Cx47 membrane localization. Inflammatory changes and connexin alterations were most severe in the KO T55I group. To examine why the presence of the T55I mutant exacerbates pathology even more than in Cx32 KO mice, we analyzed the expression of ER-stress markers BiP, Fas and CHOP by immunostaining, immunoblot and Real-time PCR. All markers were increased in LPS treated KO T55I mice more than in other genotypes. In conclusion, LPS induced neuroinflammation causes disruption of the main astrocyte-oligodendrocyte GJs, which may contribute to the increased sensitivity of Cx32 KO mice to LPS and of patients with CMT1X to various stressors. Moreover the presence of an intracellularly retained, misfolded CMT1X mutant such as T55I induces ER stress under inflammatory conditions, further exacerbating oligodendrocyte dysfunction and pathological changes in the CNS.
Collapse
Affiliation(s)
- Margarita Olympiou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Kyriaki Markoullis
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christos Karaiskos
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Alexia Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Styliana Kyriakoudi
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Charles K Abrams
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, USA
| | - Kleopas A Kleopa
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus.
- Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, 6 International Airport Avenue, P.O. Box 23462, , 1683, Nicosia, Cyprus.
| |
Collapse
|
31
|
Lunn M, Hanna M, Howard R, Parton M, Rahman S, Reilly M, Sidle K, Turner C. Nerve and Muscle Disease. Neurology 2016. [DOI: 10.1002/9781118486160.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
| | | | | | | | | | | | - Katie Sidle
- National Hospital for Neurology & Neurosurgery
| | | |
Collapse
|
32
|
Sun B, Chen ZH, Ling L, Li YF, Liu LZ, Yang F, Huang XS. Mutation Analysis of Gap Junction Protein Beta 1 and Genotype-Phenotype Correlation in X-linked Charcot-Marie-Tooth Disease in Chinese Patients. Chin Med J (Engl) 2016; 129:1011-6. [PMID: 27098783 PMCID: PMC4852665 DOI: 10.4103/0366-6999.180511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Among patients with Charcot-Marie-Tooth disease (CMT), the X-linked variant (CMTX) caused by gap junction protein beta 1 (GJB1) gene mutation is the second most frequent type, accounting for approximately 90% of all CMTX. More than 400 mutations have been identified in the GJB1 gene that encodes connexin 32 (CX32). CX32 is thought to form gap junctions that promote the diffusion pathway between cells. GJB1 mutations interfere with the formation of the functional channel and impair the maintenance of peripheral myelin, and novel mutations are continually discovered. METHODS We included 79 unrelated patients clinically diagnosed with CMT at the Department of Neurology of the Chinese People's Liberation Army General Hospital from December 20, 2012, to December 31, 2015. Clinical examination, nerve conduction studies, and molecular and bioinformatics analyses were performed to identify patients with CMTX1. RESULTS Nine GJB1 mutations (c.283G>A, c.77C>T, c.643C>T, c.515C>T, c.191G>A, c.610C>T, c.490C>T, c.491G>A, and c.44G>A) were discovered in nine patients. Median motor nerve conduction velocities of all nine patients were < 38 m/s, resembling CMT Type 1. Three novel mutations, c.643C>T, c.191G>A, and c.610C>T, were revealed and bioinformatics analyses indicated high pathogenicity. CONCLUSIONS The three novel missense mutations within the GJB1 gene broaden the mutational diversity of CMT1X. Molecular analysis of family members and bioinformatics analyses of the afflicted patients confirmed the pathogenicity of these mutations.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Zhao-Hui Chen
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Li Ling
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Yi-Fan Li
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Li-Zhi Liu
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Fei Yang
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xu-Sheng Huang
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| |
Collapse
|
33
|
Intrathecal gene therapy rescues a model of demyelinating peripheral neuropathy. Proc Natl Acad Sci U S A 2016; 113:E2421-9. [PMID: 27035961 DOI: 10.1073/pnas.1522202113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Inherited demyelinating peripheral neuropathies are progressive incurable diseases without effective treatment. To develop a gene therapy approach targeting myelinating Schwann cells that can be translatable, we delivered a lentiviral vector using a single lumbar intrathecal injection and a myelin-specific promoter. The human gene of interest, GJB1, which is mutated in X-linked Charcot-Marie-Tooth Disease (CMT1X), was delivered intrathecally into adult Gjb1-null mice, a genetically authentic model of CMT1X that develops a demyelinating peripheral neuropathy. We obtained widespread, stable, and cell-specific expression of connexin32 in up to 50% of Schwann cells in multiple lumbar spinal roots and peripheral nerves. Behavioral and electrophysiological analysis revealed significantly improved motor performance, quadriceps muscle contractility, and sciatic nerve conduction velocities. Furthermore, treated mice exhibited reduced numbers of demyelinated and remyelinated fibers and fewer inflammatory cells in lumbar motor roots, as well as in the femoral motor and sciatic nerves. This study demonstrates that a single intrathecal lentiviral gene delivery can lead to Schwann cell-specific expression in spinal roots extending to multiple peripheral nerves. This clinically relevant approach improves the phenotype of an inherited neuropathy mouse model and provides proof of principle for treating inherited demyelinating neuropathies.
Collapse
|
34
|
Wasseff SK, Scherer SS. Activated immune response in an inherited leukodystrophy disease caused by the loss of oligodendrocyte gap junctions. Neurobiol Dis 2015; 82:86-98. [PMID: 26051537 PMCID: PMC4640986 DOI: 10.1016/j.nbd.2015.05.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/30/2015] [Accepted: 05/27/2015] [Indexed: 01/11/2023] Open
Abstract
Oligodendrocyte:oligodendrocyte (O:O) gap junction (GJ) coupling is a widespread and essential feature of the CNS, and is mediated by connexin47 (Cx47) and Cx32. Loss of function mutations affecting Cx47 results in a severe leukodystrophy, Pelizeus-Merzbacher-like disease (also known as Hypomyelinating Leukodystrophy 2), which can be reproduced in mice lacking both Cx47 and Cx32. Here we report the gene expression profile of the cerebellum--an affected brain region--in mice lacking both Cx47 and Cx32. Of the 43,174 mRNA probes examined, we find decreased expression of 23 probes (corresponding to 23 genes) and increased expression of 545 probes (corresponding to 348 genes). Many of the genes with reduced expression map to oligodendrocytes, and two of them (Fa2h and Ugt8a) are involved in the synthesis of myelin lipids. Many of the genes with increased expression map to lymphocytes and microglia, and involved in leukotrienes/prostaglandins synthesis and chemokines/cytokines interactions and signaling pathways. In accord, immunostaining showed T- and B-cells in the cerebella of mutant mice as well as activated microglia and astrocytes. Thus, in addition to the loss of GJ coupling, there is a prominent immune response in mice lacking both Cx47 and Cx32.
Collapse
Affiliation(s)
- Sameh K Wasseff
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 450 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA USA 19104-6077.
| | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 450 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA USA 19104-6077.
| |
Collapse
|
35
|
Exome Sequence Analysis Suggests that Genetic Burden Contributes to Phenotypic Variability and Complex Neuropathy. Cell Rep 2015; 12:1169-83. [PMID: 26257172 DOI: 10.1016/j.celrep.2015.07.023] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 05/27/2015] [Accepted: 07/09/2015] [Indexed: 02/08/2023] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous distal symmetric polyneuropathy. Whole-exome sequencing (WES) of 40 individuals from 37 unrelated families with CMT-like peripheral neuropathy refractory to molecular diagnosis identified apparent causal mutations in ∼ 45% (17/37) of families. Three candidate disease genes are proposed, supported by a combination of genetic and in vivo studies. Aggregate analysis of mutation data revealed a significantly increased number of rare variants across 58 neuropathy-associated genes in subjects versus controls, confirmed in a second ethnically discrete neuropathy cohort, suggesting that mutation burden potentially contributes to phenotypic variability. Neuropathy genes shown to have highly penetrant Mendelizing variants (HPMVs) and implicated by burden in families were shown to interact genetically in a zebrafish assay exacerbating the phenotype established by the suppression of single genes. Our findings suggest that the combinatorial effect of rare variants contributes to disease burden and variable expressivity.
Collapse
|
36
|
Sargiannidou I, Kagiava A, Bashiardes S, Richter J, Christodoulou C, Scherer SS, Kleopa KA. Intraneural GJB1 gene delivery improves nerve pathology in a model of X-linked Charcot-Marie-Tooth disease. Ann Neurol 2015; 78:303-16. [PMID: 26010264 DOI: 10.1002/ana.24441] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE X-linked Charcot-Marie-Tooth disease (CMT1X) is a common inherited neuropathy caused by mutations in the GJB1 gene encoding the gap junction protein connexin32 (Cx32). Clinical studies and disease models indicate that neuropathy mainly results from Schwann cell autonomous, loss-of-function mechanisms; therefore, CMT1X may be treatable by gene replacement. METHODS A lentiviral vector LV.Mpz-GJB1 carrying the GJB1 gene under the Schwann cell-specific myelin protein zero (Mpz) promoter was generated and delivered into the mouse sciatic nerve by a single injection immediately distal to the sciatic notch. Enhanced green fluorescent protein (EGFP) reporter gene expression was quantified and Cx32 expression was examined on a Cx32 knockout (KO) background. A gene therapy trial was performed in a Cx32 KO model of CMT1X. RESULTS EGFP was expressed throughout the length of the sciatic nerve in up to 50% of Schwann cells starting 2 weeks after injection and remaining stable for up to 16 weeks. Following LV.Mpz-GJB1 injection into Cx32 KO nerves, we detected Cx32 expression and correct localization in non-compact myelin areas where gap junctions are normally formed. Gene therapy trial by intraneural injection in groups of 2-month-old Cx32 KO mice, before demyelination onset, significantly reduced the ratio of abnormally myelinated fibers (p = 0.00148) and secondary inflammation (p = 0.0178) at 6 months of age compared to mock-treated animals. INTERPRETATION Gene delivery using a lentiviral vector leads to efficient gene expression specifically in Schwann cells. Restoration of Cx32 expression ameliorates nerve pathology in a disease model and provides a promising approach for future treatments of CMT1X and other inherited neuropathies.
Collapse
Affiliation(s)
| | | | - Stavros Bashiardes
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Jan Richter
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christina Christodoulou
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Steven S Scherer
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Kleopas A Kleopa
- Neuroscience Laboratory
- Neurology Clinics, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| |
Collapse
|
37
|
Connexins, gap junctions and peripheral neuropathy. Neurosci Lett 2015; 596:27-32. [DOI: 10.1016/j.neulet.2014.10.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 11/23/2022]
|
38
|
Rossor AM, Evans MRB, Reilly MM. A practical approach to the genetic neuropathies. Pract Neurol 2015; 15:187-98. [DOI: 10.1136/practneurol-2015-001095] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 11/04/2022]
|
39
|
Relapsing remitting multiple sclerosis in x-linked charcot-marie-tooth disease with central nervous system involvement. Case Rep Neurol Med 2015; 2015:841897. [PMID: 25883816 PMCID: PMC4390163 DOI: 10.1155/2015/841897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/18/2015] [Indexed: 11/28/2022] Open
Abstract
We report a patient with relapsing remitting multiple sclerosis (MS) and X-linked Charcot-Marie-Tooth disease (CMTX), carrying a GJB1 mutation affecting connexin-32 (c.191G>A, p. Cys64Tyr) which was recently reported by our group. This is the third case report of a patient with CMTX developing MS, but it is unique in the fact that other family members carrying the same mutation were found to have asymptomatic central nervous system (CNS) involvement (diffuse white matter hyperintensity on brain MRI and extensor plantars). Although this may be a chance association, the increasing number of cases with CMTX and MS, especially with mutations involving the CNS, may imply some causative effect and provide insights into MS pathogenesis.
Collapse
|
40
|
Xiao F, Tan JZ, Zhang X, Wang XF. A novel mutation in GJB1 (c.212T>G) in a Chinese family with X-linked Charcot-Marie-Tooth disease. J Clin Neurosci 2015; 22:513-8. [PMID: 25595958 DOI: 10.1016/j.jocn.2014.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/14/2014] [Accepted: 08/04/2014] [Indexed: 11/16/2022]
Abstract
Gap junction protein beta 1 (GJB1) gene mutations lead to X-linked Charcot-Marie-Tooth (CMTX) disease. We investigated a Chinese family with CMTX and identified a novel GJB1 point mutation. Clinical and electrophysiological features of the pedigree were examined, and sequence alterations of the coding region of GJB1 that encode connexin32 were determined by direct sequencing. Sequence alignment of the mutation site was performed using Clustal W. Mutation effects were analysed using PolyPhen-2, SIFT and Mutation Taster software. The three-dimensional structures of the mutant and wild-type proteins were predicted by modeling with SWISS MODEL online software. The affected family members displayed typical Charcot-Marie-Tooth phenotypes, but phenotypic heterogeneity was observed. Nerve conduction velocities of all affected patients were slow. Sequencing of GJB1 revealed a heterozygous T>G missense mutation at nucleotide 212 in the proband, the proband's mother and the proband's daughter. The affected male sibling of the proband displayed a hemizygous missense mutation with T>G transition at the identical position on the GJB1 gene. This mutation resulted in an amino acid change from isoleucine to serine that was predicted to lead to tertiary structural alterations that would disrupt the function of the GJB1 protein. A novel point mutation in GJB1 was detected, expanding the spectrum of GJB1 mutations known to be associated with CMTX.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 You Yi Road, Chongqing 400016, China
| | - Jia-ze Tan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 You Yi Road, Chongqing 400016, China
| | - Xu Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 You Yi Road, Chongqing 400016, China
| | - Xue-Feng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 You Yi Road, Chongqing 400016, China.
| |
Collapse
|
41
|
Mathis S, Magy L, Vallat JM. Therapeutic options in Charcot–Marie–Tooth diseases. Expert Rev Neurother 2015; 15:355-66. [DOI: 10.1586/14737175.2015.1017471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Bouhy D, Timmerman V. Animal models and therapeutic prospects for Charcot-Marie-Tooth disease. Ann Neurol 2013; 74:391-6. [PMID: 23913540 DOI: 10.1002/ana.23987] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/04/2013] [Accepted: 07/29/2013] [Indexed: 12/14/2022]
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are inherited neuromuscular disorders caused by a length-dependent neurodegeneration of peripheral nerves. More than 900 mutations in 60 different genes are causative of the neuropathy. Despite significant progress in therapeutic strategies, the disease remains incurable. The increasing number of genes linked to the disease, and their considerable clinical and genetic heterogeneity render the development of these strategies particularly challenging. In this context, cellular and animals models provide powerful tools. Efficient motor and sensory tests have been developed to assess the behavioral phenotype in transgenic animal models (rodent and fly). When these models reproduce a phenotype comparable to CMT, they allow therapeutic approaches and the discovery of modifiers and biomarkers. In this review, we describe the most convincing transgenic rodent and fly models of CMT and how they can lead to clinical trial. We also discuss the challenges that the research, the clinic, and the pharmaceutical industry will face in developing efficient and accessible treatment for CMT patients.
Collapse
Affiliation(s)
- Delphine Bouhy
- Peripheral Neuropathy Group, Department of Molecular Genetics, Flanders Interuniversity Institute for Biotechnology, Institute Born Bunge, University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
43
|
Bouhy D, Timmerman V. Modèles animaux dans la maladie de Charcot-Marie-Tooth et applications de la compréhension de la maladie chez l’homme. Rev Neurol (Paris) 2013; 169:971-7. [DOI: 10.1016/j.neurol.2013.07.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 11/26/2022]
|
44
|
Borgulová I, Mazanec R, Sakmaryová I, Havlová M, Safka Brožková D, Seeman P. Mosaicism for GJB1 mutation causes milder Charcot-Marie-Tooth X1 phenotype in a heterozygous man than in a manifesting heterozygous woman. Neurogenetics 2013; 14:189-95. [PMID: 23912496 DOI: 10.1007/s10048-013-0368-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/19/2013] [Indexed: 01/11/2023]
Abstract
Charcot-Marie-Tooth (CMT) disease is a heterogeneous disorder of the peripheral nervous system that collectively affects approximately 1 in 2,500 individuals, thus making it the most common inherited neurologic disorder. X-linked inheritance may account for 10-20 % of CMT neuropathy. We report a Czech family with a 30-year-old woman affected by CMT since the age of 10 years, originally as an isolated case. Nerve conduction study (NCS) showed demyelinating neuropathy, and DNA testing revealed a novel heterozygous gap junction beta-1 protein (GJB1) mutation c.784_786delTA. The same mutation, but surprisingly in heterozygous state, was subsequently found in her subjectively healthy father and later also in one of her sisters but not in her two other sisters. NCS showed intermediate type of motor and sensory neuropathy in these two females manifesting heterozygotes and normal results in the other healthy sisters and one brother, all without the c.784_786delTA mutation. The father has a phenotype milder than his daughter and has only subclinical signs of CMT. The index female patient had normal karyotype 46, XX, and normal FISH for centromeric X chromosome. We concluded that the proband's father is a heterozygote due to the somatic mosaicism for the GJB1 mutation in his leukocytes (detected by DNA sequencing) and also in his germ cells as confirmed by the unexpectedly different genotypes in his four daughters. Quantitative analysis revealed a mutated signal in 25:75 allele proportion of mutated to healthy allele in the mosaic father. This study has important consequences for genetic counseling and prognosis in CMTX1 families.
Collapse
Affiliation(s)
- I Borgulová
- Centre for Medical Genetics and Reproductive Medicine GENNET, Prague, Czech Republic,
| | | | | | | | | | | |
Collapse
|
45
|
Morrison BM, Lee Y, Rothstein JD. Oligodendroglia: metabolic supporters of axons. Trends Cell Biol 2013; 23:644-51. [PMID: 23988427 DOI: 10.1016/j.tcb.2013.07.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 11/19/2022]
Abstract
Axons are specialized extensions of neurons that are critical for the organization of the nervous system. To maintain function in axons that often extend some distance from the cell body, specialized mechanisms of energy delivery are likely to be necessary. Over the past decade, greater understanding of human demyelinating diseases and the development of animal models have suggested that oligodendroglia are critical for maintaining the function of axons. In this review, we discuss evidence for the vulnerability of neurons to energy deprivation, the importance of oligodendrocytes for axon function and survival, and recent data suggesting that transfer of energy metabolites from oligodendroglia to axons through monocarboxylate transporter 1 (MCT1) may be critical for the survival of axons. This pathway has important implications both for the basic biology of the nervous system and for human neurological disease. New insights into the role of oligodendroglial biology provide an exciting opportunity for revisions in nervous system biology, understanding myelin-based disorders, and therapeutics development.
Collapse
Affiliation(s)
- Brett M Morrison
- Brain Science Institute, Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
46
|
Gerding WM, Koetting J, Rey LP, Bibas Bonet H, Abdala ME, Mazzeo A, Mostacciuolo ML, Arning L, Carrero-Valenzuela R. Hereditary motor and sensory neuropathy (HMSN) type X1 in an Argentinean family reveals independent GJB1/Cx32 mutations at the identical nucleotide position. Mol Cell Probes 2013; 27:118-21. [PMID: 23384994 DOI: 10.1016/j.mcp.2013.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/04/2012] [Accepted: 01/18/2013] [Indexed: 11/21/2022]
Abstract
X-linked Charcot-Marie-Tooth disease (CMT Type X1, OMIM: 302800) represents a frequent cause of hereditary peripheral motor and sensory neuropathies and is associated with mutations in GJB1 encoding the gap junction beta 1 protein connexin 32 (Cx32). Studying an Argentinean family of Italian origin with seven affected males in three generations exhibiting clinical signs of CMT, eight obligate female carriers were identified genealogically. DNA sequencing of exon 2 and adjacent regions of the GJB1 gene in two symptomatic males whose respective maternal grandfathers, both affected, were brothers, revealed mutations in GJB1/Cx32. Surprisingly, each of the two affected patients had a different mutation in hemizygous state at the same nucleotide position: c.383C>T (p.S128L) and c.383C>A (p.S128X). In both cases, the identified mutation was present in heterozygous state in the corresponding maternal genomic DNA. Furthermore, X-chromosomal microsatellite analysis showed identical marker alleles in both patients. Together with the genealogical information, these molecular data imply that a primarily mutated allele mutated for a second time. In conclusion, two different mutations at the same nucleotide position in this Argentinean family represent a finding with a very low probability of occurrence.
Collapse
|
47
|
Chaudhry R, Kidambi A, Brewer MH, Antonellis A, Mathews K, Nicholson G, Kennerson M. Re-analysis of an original CMTX3 family using exome sequencing identifies a known BSCL2 mutation. Muscle Nerve 2013; 47:922-4. [PMID: 23553728 DOI: 10.1002/mus.23743] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 11/25/2012] [Accepted: 11/27/2012] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Charcot-Marie-Tooth (CMT) disease is a group of peripheral neuropathies affecting both motor and sensory nerves. CMTX3 is an X-linked CMT locus, which maps to chromosome Xq26.3-q27.3. Initially, CMTX3 was mapped to a 31.2-Mb region in 2 American families. We have reexamined 1 of the original families (US-PED2) by next generation sequencing. METHODS Three members of the family underwent exome sequencing. Candidate variants were validated by PCR and Sanger sequencing analysis. CONCLUSION No pathogenic coding variants localizing to the CMTX3 region were identified. However, exome sequencing identified a known BSCL2 mutation (N88S). This study demonstrates the power of exome sequencing as a tool to identify gene mutations for a small family in the absence of statistically significant linkage data.
Collapse
Affiliation(s)
- Rabia Chaudhry
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Gate 3, Hospital Road, Concord, New South Wales, 2761, Australia.
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Hereditary neuropathies (HN) with onset in childhood are categorized according to clinical presentation, pathogenic mechanism based on electrophysiology, genetic transmission and, in selected cases, pathological findings. Especially relevant to pediatrics are the items "secondary" versus "primary" neuropathy, "syndromic versus nonsyndromic," and "period of life." Different combinations of these parameters frequently point toward specific monogenic disorders. Ruling out a neuropathy secondary to a generalized metabolic disorder remains the first concern in pediatrics. As a rule, metabolic diseases include additional, orienting symptoms or signs, and their biochemical diagnosis is based on logical algorithms. Primary, motor sensory are the most frequent HN and are dominated by demyelinating autosomal dominant (AD) forms (CMT1). Other forms include demyelinating autosomal recessive (AR) forms, axonal AD/AR forms, and forms with "intermediate" electrophysiological phenotype. Peripheral motor neuron disorders are dominated by AR SMN-linked spinal muscular atrophies. (Distal) hereditary motor neuropathies represent <10% of HN but exhibit large clinical and genetic heterogeneity. Sensory/dysautonomic HN involves five classic subtypes, each one related to specific genes. However, genetic heterogeneity is larger than initially suspected. Syndromic HN distinguish "purely neurological syndromes", which are multisystemic, such as spinocerebellar atrophies +, spastic paraplegias +, etc. Peripheral neuropathy is possibly the presenting feature, including in childhood. Autosomal recessive forms, on average, start more frequently in childhood. "Multiorgan syndromes", on the other hand, are more specific to Pediatrics. AR forms, which are clearly degenerative, prompt the investigation of a large set of pleiotropic genes. Other syndromes expressed in the perinatal period are mainly developmental disorders, and can sometimes be related to specific transcription factors. Systematic malformative workup and ethical considerations are necessary. Altogether, >40 genes with various biological functions have been found to be responsible for primary HN. Many are responsible for various phenotypes, including some without the polyneuropathic trait, and some for various types of transmission.
Collapse
Affiliation(s)
- Pierre Landrieu
- Department of Pediatric Neurology, CHU Paris sud, Hôpital Bicêtre, Paris, France.
| | | |
Collapse
|
49
|
Landrieu P, Baets J, De Jonghe P. Hereditary motor-sensory, motor, and sensory neuropathies in childhood. HANDBOOK OF CLINICAL NEUROLOGY 2013; 113:1413-32. [PMID: 23622364 DOI: 10.1016/b978-0-444-59565-2.00011-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hereditary neuropathies (HN) are categorized according to clinical presentation, pathogenic mechanism based on electrophysiology, genetic transmission, age of occurrence, and, in selected cases, pathological findings. The combination of these parameters frequently orients towards specific genetic disorders. Ruling out a neuropathy secondary to a generalized metabolic disorder remains the first pediatric concern. Primary, motor-sensory are the most frequent HN and are dominated by demyelinating AD forms (CMT1). Others are demyelinating AR forms, axonal AD/AR forms, and forms with "intermediate" electrophysiological phenotype. Pure motor HN represent<10% of HN but exhibit large clinical and genetic heterogeneity. Sensory/dysautonomic HN cover five classical subtypes, each one related to specific genes. However, genetic heterogeneity is largly greater than initially suspected. Syndromic HN distinguish: "purely neurological syndromes", which are multisystemic, usually AD disorders, such as spinocerebellar atrophies +, spastic paraplegias +, etc. Peripheral Neuropathy may be the presenting feature, including in childhood. Clearly degenerative, AR forms prompt to investigate a large set of pleiotropic genes. Other syndromes, expressed in the perinatal period and comprising malformative features, are mainly developmental disorders, sometimes related to specific transcription factors. Altogether, >40 genes with various biological functions have been found responsible for HN. Many are responsible for various phenotypes, including some without the polyneuropathic trait: for the pediatric neurologist, phenotype/genotype correlations constitute a permanent bidirectional exercise.
Collapse
Affiliation(s)
- Pierre Landrieu
- Department of Paediatric Neurology, Université Paris Sud, Bicêtre Hospital, Paris, France.
| | | | | |
Collapse
|
50
|
Abstract
The inherited neuropathies are a clinically and genetically heterogeneous group of disorders in which there have been rapid advances in the last two decades. Molecular genetic testing is now an integral part of the evaluation of patients with inherited neuropathies. In this chapter we describe the genes responsible for the primary inherited neuropathies. We briefly discuss the clinical phenotype of each of the known inherited neuropathy subgroups, describe algorithms for molecular genetic testing of affected patients and discuss genetic counseling. The basic principles of careful phenotyping, documenting an accurate family history, and testing the available genes in an appropriate manner should identify the vast majority of individuals with CMT1 and many of those with CMT2. In this chapter we also describe the current methods of genetic testing. As advances are made in molecular genetic technologies and improvements are made in bioinformatics, it is likely that the current time-consuming methods of DNA sequencing will give way to quicker and more efficient high-throughput methods, which are briefly discussed here.
Collapse
|