1
|
Parmar JM, Laing NG, Kennerson ML, Ravenscroft G. Genetics of inherited peripheral neuropathies and the next frontier: looking backwards to progress forwards. J Neurol Neurosurg Psychiatry 2024; 95:992-1001. [PMID: 38744462 PMCID: PMC11503175 DOI: 10.1136/jnnp-2024-333436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Inherited peripheral neuropathies (IPNs) encompass a clinically and genetically heterogeneous group of disorders causing length-dependent degeneration of peripheral autonomic, motor and/or sensory nerves. Despite gold-standard diagnostic testing for pathogenic variants in over 100 known associated genes, many patients with IPN remain genetically unsolved. Providing patients with a diagnosis is critical for reducing their 'diagnostic odyssey', improving clinical care, and for informed genetic counselling. The last decade of massively parallel sequencing technologies has seen a rapid increase in the number of newly described IPN-associated gene variants contributing to IPN pathogenesis. However, the scarcity of additional families and functional data supporting variants in potential novel genes is prolonging patient diagnostic uncertainty and contributing to the missing heritability of IPNs. We review the last decade of IPN disease gene discovery to highlight novel genes, structural variation and short tandem repeat expansions contributing to IPN pathogenesis. From the lessons learnt, we provide our vision for IPN research as we anticipate the future, providing examples of emerging technologies, resources and tools that we propose that will expedite the genetic diagnosis of unsolved IPN families.
Collapse
Affiliation(s)
- Jevin M Parmar
- Rare Disease Genetics and Functional Genomics, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Nigel G Laing
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Preventive Genetics, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, New South Wales, Australia
- Molecular Medicine Laboratory, Concord Hospital, Concord, New South Wales, Australia
| | - Gianina Ravenscroft
- Rare Disease Genetics and Functional Genomics, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
2
|
Prior R, Silva A, Vangansewinkel T, Idkowiak J, Tharkeshwar AK, Hellings TP, Michailidou I, Vreijling J, Loos M, Koopmans B, Vlek N, Agaser C, Kuipers TB, Michiels C, Rossaert E, Verschoren S, Vermeire W, de Laat V, Dehairs J, Eggermont K, van den Biggelaar D, Bademosi AT, Meunier FA, vandeVen M, Van Damme P, Mei H, Swinnen JV, Lambrichts I, Baas F, Fluiter K, Wolfs E, Van Den Bosch L. PMP22 duplication dysregulates lipid homeostasis and plasma membrane organization in developing human Schwann cells. Brain 2024; 147:3113-3130. [PMID: 38743588 PMCID: PMC11370802 DOI: 10.1093/brain/awae158] [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: 12/06/2023] [Revised: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited peripheral neuropathy caused by a 1.5 Mb tandem duplication of chromosome 17 harbouring the PMP22 gene. This dose-dependent overexpression of PMP22 results in disrupted Schwann cell myelination of peripheral nerves. To obtain better insights into the underlying pathogenic mechanisms in CMT1A, we investigated the role of PMP22 duplication in cellular homeostasis in CMT1A mouse models and in patient-derived induced pluripotent stem cells differentiated into Schwann cell precursors (iPSC-SCPs). We performed lipidomic profiling and bulk RNA sequencing (RNA-seq) on sciatic nerves of two developing CMT1A mouse models and on CMT1A patient-derived iPSC-SCPs. For the sciatic nerves of the CMT1A mice, cholesterol and lipid metabolism was downregulated in a dose-dependent manner throughout development. For the CMT1A iPSC-SCPs, transcriptional analysis unveiled a strong suppression of genes related to autophagy and lipid metabolism. Gene ontology enrichment analysis identified disturbances in pathways related to plasma membrane components and cell receptor signalling. Lipidomic analysis confirmed the severe dysregulation in plasma membrane lipids, particularly sphingolipids, in CMT1A iPSC-SCPs. Furthermore, we identified reduced lipid raft dynamics, disturbed plasma membrane fluidity and impaired cholesterol incorporation and storage, all of which could result from altered lipid storage homeostasis in the patient-derived CMT1A iPSC-SCPs. Importantly, this phenotype could be rescued by stimulating autophagy and lipolysis. We conclude that PMP22 duplication disturbs intracellular lipid storage and leads to a more disordered plasma membrane owing to an alteration in the lipid composition, which might ultimately lead to impaired axo-glial interactions. Moreover, targeting lipid handling and metabolism could hold promise for the treatment of patients with CMT1A.
Collapse
Affiliation(s)
- Robert Prior
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn 53127, Germany
| | - Alessio Silva
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Tim Vangansewinkel
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
- UHasselt—Hasselt University, Biomedical Research Institute, Diepenbeek 3590, Belgium
| | - Jakub Idkowiak
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven 3000, Belgium
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Pardubice 532 10, Czech Republic
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Tom P Hellings
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Iliana Michailidou
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Jeroen Vreijling
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Maarten Loos
- InnoSer Nederland B.V., 2333 CK Leiden, The Netherlands
| | | | - Nina Vlek
- InnoSer Nederland B.V., 2333 CK Leiden, The Netherlands
| | - Cedrick Agaser
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Thomas B Kuipers
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Christine Michiels
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Elisabeth Rossaert
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Stijn Verschoren
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Wendy Vermeire
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Vincent de Laat
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Kristel Eggermont
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Diede van den Biggelaar
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| | - Adekunle T Bademosi
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frederic A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Martin vandeVen
- UHasselt—Hasselt University, Biomedical Research Institute, Diepenbeek 3590, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven 3000, Belgium
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Ivo Lambrichts
- UHasselt—Hasselt University, Biomedical Research Institute, Diepenbeek 3590, Belgium
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Esther Wolfs
- UHasselt—Hasselt University, Biomedical Research Institute, Diepenbeek 3590, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven 3000, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven 3000, Belgium
| |
Collapse
|
3
|
van Eyll J, Prior R, Celanire S, Van Den Bosch L, Rombouts F. Therapeutic indications for HDAC6 inhibitors in the peripheral and central nervous disorders. Expert Opin Ther Targets 2024; 28:719-737. [PMID: 39305025 DOI: 10.1080/14728222.2024.2404571] [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: 04/16/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Inhibition of the enzymatic function of HDAC6 is currently being explored in clinical trials ranging from peripheral neuropathies to cancers. Advances in selective HDAC6 inhibitor discovery allowed studying highly efficacious brain penetrant and peripheral restrictive compounds for treating PNS and CNS indications. AREAS COVERED This review explores the multifactorial role of HDAC6 in cells, the common pathological hallmarks of PNS and CNS disorders, and how HDAC6 modulates these mechanisms. Pharmacological inhibition of HDAC6 and genetic knockout/knockdown studies as a therapeutic strategy in PNS and CNS indications were analyzed. Furthermore, we describe the recent developments in HDAC6 PET tracers and their utility in CNS indications. Finally, we explore the advancements and challenges with HDAC6 inhibitor compounds, such as hydroxamic acid, fluoromethyl oxadiazoles, HDAC6 degraders, and thiol-based inhibitors. EXPERT OPINION Based on extensive preclinical evidence, pharmacological inhibition of HDAC6 is a promising approach for treating both PNS and CNS disorders, given its involvement in neurodegeneration and aging-related cellular processes. Despite the progress in the development of selective HDAC6 inhibitors, safety concerns remain regarding their chronic administration in PNS and CNS indications, and the development of novel compound classes and modalities inhibiting HDAC6 function offer a way to mitigate some of these safety concerns.
Collapse
Affiliation(s)
| | | | - Sylvain Celanire
- Augustine Therapeutics, Research and Development, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | | |
Collapse
|
4
|
Mills JF, Heiland LD, Nguyen SA, Close MF, Meyer TA. Charcot-Marie-Tooth Disease and Hearing Loss: A Systematic Review With Meta-Analysis. Otol Neurotol 2024; 45:732-739. [PMID: 38956759 DOI: 10.1097/mao.0000000000004243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
OBJECTIVE To characterize the pattern of hearing loss in Charcot-Marie-Tooth (CMT) disease to help guide clinical management. DATABASES REVIEWED CINAHL, PubMed, and Scopus. METHODS Two independent investigators selected studies on CMT patients with pure-tone average (PTA) and auditory brainstem response (ABR) data. Case reports, case series <5 patients, and data that overlapped with another study were excluded. Investigators performed data extraction, quality rating, and risk-of-bias assessment using the Newcastle-Ottawa Scale. Meta-analysis of mean difference using fixed/random effects models was used. Also, data were analyzed using a weighted one-way analysis of variance, with post-hoc Tukey's test for comparison. RESULTS Ultimately, 6 prospective studies (N = 197) were included. The most common demyelinating subtype (CMT1A) had significantly prolonged ABR latency values across wave III (0.20 ms, 95% confidence interval [CI]: 0.05-0.35), wave V (0.20 ms, 95% CI: 0.01-0.39), waves I-III (0.20 ms, 95% CI: 0.01-0.39), and waves I-V (0.20 ms, 95% CI: 0.01-0.39) when compared to matched controls. The autosomal recessive demyelinating subtype (CMT4C) had significantly worse PTA when compared to the most common subtype (CMT1A) (Δ 28.93 dB, 95% CI 18.34-39.52) and nondemyelinating subtype (CMT2A) (Δ 28.3 dB, 95% CI: 15.98-40.62). CONCLUSIONS Patients with CMT can present with a variety of phenotypes depending on the causative mutation. The ABR interpeak latency values for the most common demyelinating form of CMT are delayed when compared to matched controls. Most subtypes have normal hearing thresholds, apart from CMT4C, which presents with mild hearing loss on average.
Collapse
Affiliation(s)
| | - Luke D Heiland
- Department of Otolaryngology, Medical University of South Carolina, South Carolina
| | - Shaun A Nguyen
- Department of Otolaryngology, Medical University of South Carolina, South Carolina
| | - Michaela F Close
- Department of Otolaryngology, Medical University of South Carolina, South Carolina
| | - Ted A Meyer
- Department of Otolaryngology, Medical University of South Carolina, South Carolina
| |
Collapse
|
5
|
Doherty CM, Howard P, O'Donnell LF, Zuccarino R, Wastling S, Milev E, Banks T, Shah S, Zafeiropoulos N, Stephens KJ, Sarkozy A, Grider T, Feely SME, Manzur A, Shy RR, Skorupinska M, Pipis M, Nicolaisen E, McDowell A, Dilek N, Rossor AM, Laura M, Clark C, Muntoni F, Thedens D, Thornton J, Morrow JM, Shy ME, Reilly MM. Quantitative Foot Muscle Magnetic Resonance Imaging Reliably Measures Disease Progression in Children and Adolescents with Charcot-Marie-Tooth Disease Type 1A. Ann Neurol 2024; 96:170-174. [PMID: 38613459 DOI: 10.1002/ana.26934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Quantitative muscle fat fraction (FF) responsiveness is lower in younger Charcot-Marie-Tooth disease type 1A (CMT1A) patients with lower baseline calf-level FF. We investigated the practicality, validity, and responsiveness of foot-level FF in this cohort involving 22 CMT1A patients and 14 controls. The mean baseline foot-level FF was 25.9 ± 20.3% in CMT1A patients, and the 365-day FF (n = 15) increased by 2.0 ± 2.4% (p < 0.001 vs controls). Intrinsic foot-level FF demonstrated large responsiveness (12-month standardized response mean (SRM) of 0.86) and correlated with the CMT examination score (ρ = 0.58, P = 0.01). Intrinsic foot-level FF has the potential to be used as a biomarker in future clinical trials involving younger CMT1A patients. ANN NEUROL 2024;96:170-174.
Collapse
Affiliation(s)
- Carolynne M Doherty
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Paige Howard
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Luke F O'Donnell
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Riccardo Zuccarino
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Fondazione Serena Onlus, Centro Clinico NeMO Trento, Italy
| | - Stephen Wastling
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Evelin Milev
- Dubowitz Neuromuscular Center, Great Ormond Street Hospital, London, UK
| | - Tina Banks
- Department of Radiology, Great Ormond Street Hospital, London, UK
| | - Sachit Shah
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Nick Zafeiropoulos
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Katherine J Stephens
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Anna Sarkozy
- Dubowitz Neuromuscular Center, Great Ormond Street Hospital, London, UK
| | - Tiffany Grider
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Shawna M E Feely
- Division of Pediatric Neurology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, USA
| | - Adnan Manzur
- Dubowitz Neuromuscular Center, Great Ormond Street Hospital, London, UK
| | - Rosemary R Shy
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mariola Skorupinska
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Menelaos Pipis
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Emma Nicolaisen
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy McDowell
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Nuran Dilek
- University of Rochester School of Medicine and Dentistry, New York, NY, USA
| | - Alexander M Rossor
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Matilde Laura
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Center, Great Ormond Street Hospital, London, UK
| | - Daniel Thedens
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John Thornton
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Jasper M Morrow
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Michael E Shy
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mary M Reilly
- Center for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
6
|
Xu IRL, Danzi MC, Ruiz A, Raposo J, De Jesus YA, Reilly MM, Cortese A, Shy ME, Scherer SS, Hermann D, Fridman V, Baets J, Saporta M, Seyedsadjadi R, Stojkovic T, Claeys KG, Patel P, Feely S, Rebelo A, Dohrn MF, Züchner S. A study concept of expeditious clinical enrollment for genetic modifier studies in Charcot-Marie-Tooth neuropathy 1A. J Peripher Nerv Syst 2024; 29:202-212. [PMID: 38581130 PMCID: PMC11209807 DOI: 10.1111/jns.12621] [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: 01/24/2024] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Caused by duplications of the gene encoding peripheral myelin protein 22 (PMP22), Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common hereditary neuropathy. Despite this shared genetic origin, there is considerable variability in clinical severity. It is hypothesized that genetic modifiers contribute to this heterogeneity, the identification of which may reveal novel therapeutic targets. In this study, we present a comprehensive analysis of clinical examination results from 1564 CMT1A patients sourced from a prospective natural history study conducted by the RDCRN-INC (Inherited Neuropathy Consortium). Our primary objective is to delineate extreme phenotype profiles (mild and severe) within this patient cohort, thereby enhancing our ability to detect genetic modifiers with large effects. METHODS We have conducted large-scale statistical analyses of the RDCRN-INC database to characterize CMT1A severity across multiple metrics. RESULTS We defined patients below the 10th (mild) and above the 90th (severe) percentiles of age-normalized disease severity based on the CMT Examination Score V2 and foot dorsiflexion strength (MRC scale). Based on extreme phenotype categories, we defined a statistically justified recruitment strategy, which we propose to use in future modifier studies. INTERPRETATION Leveraging whole genome sequencing with base pair resolution, a future genetic modifier evaluation will include single nucleotide association, gene burden tests, and structural variant analysis. The present work not only provides insight into the severity and course of CMT1A, but also elucidates the statistical foundation and practical considerations for a cost-efficient and straightforward patient enrollment strategy that we intend to conduct on additional patients recruited globally.
Collapse
Affiliation(s)
- Isaac R. L. Xu
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Matt C. Danzi
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ariel Ruiz
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Jacquelyn Raposo
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Yeisha Arcia De Jesus
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square
| | - Andrea Cortese
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square
| | - Michael E Shy
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Steven S. Scherer
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, United States
| | - David Hermann
- Department of Neurology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 673, Rochester, New York, 14642, USA
| | - Vera Fridman
- Department of Neurology, University of Colorado Anschutz Medical Campus, 12631 E 17th Avenue, Mailstop B185, Room 5113C, Aurora, CO, 80045, USA
| | - Jonathan Baets
- Department of Neurology, Neuromuscular Reference Centre, Antwerp University Hospital, Antwerp, Belgium
- Faculty of Medicine and Health Sciences, Translational Neurosciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Mario Saporta
- Department of Neurology, University of Miami Miller School of Medicine, United States
| | - Reza Seyedsadjadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tanya Stojkovic
- AP-HP, Centre de référence des maladies neuromusculaires Nord/Est/Ile de France, Hôpital Pitié-Salpêtrière, 47-83, boulevard de l’Hôpital, 75013 Paris, France
| | - Kristl G. Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Muscle Diseases and Neuropathies, KU Leuven, Leuven, Belgium
| | - Pooja Patel
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Shawna Feely
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Adriana Rebelo
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Maike F. Dohrn
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Neurology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Krauter D, Stausberg D, Hartmann TJ, Volkmann S, Kungl T, Rasche DA, Saher G, Fledrich R, Stassart RM, Nave KA, Goebbels S, Ewers D, Sereda MW. Targeting PI3K/Akt/mTOR signaling in rodent models of PMP22 gene-dosage diseases. EMBO Mol Med 2024; 16:616-640. [PMID: 38383802 PMCID: PMC10940316 DOI: 10.1038/s44321-023-00019-5] [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: 05/22/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 02/23/2024] Open
Abstract
Haplo-insufficiency of the gene encoding the myelin protein PMP22 leads to focal myelin overgrowth in the peripheral nervous system and hereditary neuropathy with liability to pressure palsies (HNPP). Conversely, duplication of PMP22 causes Charcot-Marie-Tooth disease type 1A (CMT1A), characterized by hypomyelination of medium to large caliber axons. The molecular mechanisms of abnormal myelin growth regulation by PMP22 have remained obscure. Here, we show in rodent models of HNPP and CMT1A that the PI3K/Akt/mTOR-pathway inhibiting phosphatase PTEN is correlated in abundance with PMP22 in peripheral nerves, without evidence for direct protein interactions. Indeed, treating DRG neuron/Schwann cell co-cultures from HNPP mice with PI3K/Akt/mTOR pathway inhibitors reduced focal hypermyelination. When we treated HNPP mice in vivo with the mTOR inhibitor Rapamycin, motor functions were improved, compound muscle amplitudes were increased and pathological tomacula in sciatic nerves were reduced. In contrast, we found Schwann cell dedifferentiation in CMT1A uncoupled from PI3K/Akt/mTOR, leaving partial PTEN ablation insufficient for disease amelioration. For HNPP, the development of PI3K/Akt/mTOR pathway inhibitors may be considered as the first treatment option for pressure palsies.
Collapse
Affiliation(s)
- Doris Krauter
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Daniela Stausberg
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Timon J Hartmann
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan Volkmann
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - David A Rasche
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robert Fledrich
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ruth M Stassart
- Institute of Neuropathology, University of Leipzig, Leipzig, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - David Ewers
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
| | - Michael W Sereda
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
8
|
Doherty CM, Morrow JM, Zuccarino R, Howard P, Wastling S, Pipis M, Zafeiropoulos N, Stephens KJ, Grider T, Feely SME, Nopoulous P, Skorupinska M, Milev E, Nicolaisen E, Dudzeic M, McDowell A, Dilek N, Muntoni F, Rossor AM, Shah S, Laura M, Yousry TA, Thedens D, Thornton J, Shy ME, Reilly MM. Lower limb muscle MRI fat fraction is a responsive outcome measure in CMT X1, 1B and 2A. Ann Clin Transl Neurol 2024; 11:607-617. [PMID: 38173284 DOI: 10.1002/acn3.51979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVE With potential therapies for many forms of Charcot-Marie-Tooth disease (CMT), responsive outcome measures are urgently needed for clinical trials. Quantitative lower limb MRI demonstrated progressive calf intramuscular fat accumulation in the commonest form, CMT1A with large responsiveness. In this study, we evaluated the responsiveness and validity in the three other common forms, due to variants in GJB1 (CMTX1), MPZ (CMT1B) and MFN2 (CMT2A). METHODS 22 CMTX1, 21 CMT1B and 21 CMT2A patients and matched controls were assessed at a 1-year interval. Intramuscular fat fraction (FF) was evaluated using three-point Dixon MRI at thigh and calf level along with clinical measures including CMT examination score, clinical strength assessment, CMT-HI and plasma neurofilament light chain. RESULTS All patient groups had elevated muscle fat fraction at thigh and calf levels, with highest thigh FF and atrophy in CMT2A. There was moderate correlation between calf muscle FF and clinical measures (CMTESv2 rho = 0.405; p = 0.001, ankle MRC strength rho = -0.481; p < 0.001). Significant annualised progression in calf muscle FF was seen in all patient groups (CMTX1 2.0 ± 2.0%, p < 0.001, CMT1B 1.6 ± 2.1% p = 0.004 and CMT2A 1.6 ± 2.1% p = 0.002). Greatest increase was seen in patients with 10-70% FF at baseline (calf 2.7 ± 2.3%, p < 0.0001 and thigh 1.7 ± 2.1%, p = 0.01). INTERPRETATION Our results confirm that calf muscle FF is highly responsive over 12 months in three additional common forms of CMT which together with CMT1A account for 90% of genetically confirmed cases. Calf muscle MRI FF should be a valuable outcome measure in upcoming CMT clinical trials.
Collapse
Affiliation(s)
- Carolynne M Doherty
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Jasper M Morrow
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Riccardo Zuccarino
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Fondazione Serena Onlus, Centro Clinico NeMO Trento, Pergine Valsugana, Italy
| | - Paige Howard
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Stephen Wastling
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Menelaos Pipis
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Nick Zafeiropoulos
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Katherine J Stephens
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Tiffany Grider
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shawna M E Feely
- Seattle Children's Hospital, University of Washington School of Medicine, Seattle, Washington, USA
| | - Peggy Nopoulous
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Mariola Skorupinska
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | | | - Emma Nicolaisen
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Magdalena Dudzeic
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Amy McDowell
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Nuran Dilek
- University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | - Alexander M Rossor
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Sachit Shah
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Matilde Laura
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Tarek A Yousry
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Daniel Thedens
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - John Thornton
- Lysholm Department of Radiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Michael E Shy
- Roy and Lucille Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
9
|
Bremer J, Meinhardt A, Katona I, Senderek J, Kämmerer‐Gassler EK, Roos A, Ferbert A, Schröder JM, Nikolin S, Nolte K, Sellhaus B, Popzhelyazkova K, Tacke F, Schara‐Schmidt U, Neuen‐Jacob E, de Groote CC, de Jonghe P, Timmerman V, Baets J, Weis J. Myelin protein zero mutation-related hereditary neuropathies: Neuropathological insight from a new nerve biopsy cohort. Brain Pathol 2024; 34:e13200. [PMID: 37581289 PMCID: PMC10711263 DOI: 10.1111/bpa.13200] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023] Open
Abstract
Myelin protein zero (MPZ/P0) is a major structural protein of peripheral nerve myelin. Disease-associated variants in the MPZ gene cause a wide phenotypic spectrum of inherited peripheral neuropathies. Previous nerve biopsy studies showed evidence for subtype-specific morphological features. Here, we aimed at enhancing the understanding of these subtype-specific features and pathophysiological aspects of MPZ neuropathies. We examined archival material from two Central European centers and systematically determined genetic, clinical, and neuropathological features of 21 patients with MPZ mutations compared to 16 controls. Cases were grouped based on nerve conduction data into congenital hypomyelinating neuropathy (CHN; n = 2), demyelinating Charcot-Marie-Tooth (CMT type 1; n = 11), intermediate (CMTi; n = 3), and axonal CMT (type 2; n = 5). Six cases had combined muscle and nerve biopsies and one underwent autopsy. We detected four MPZ gene variants not previously described in patients with neuropathy. Light and electron microscopy of nerve biopsies confirmed fewer myelinated fibers, more onion bulbs and reduced regeneration in demyelinating CMT1 compared to CMT2/CMTi. In addition, we observed significantly more denervated Schwann cells, more collagen pockets, fewer unmyelinated axons per Schwann cell unit and a higher density of Schwann cell nuclei in CMT1 compared to CMT2/CMTi. CHN was characterized by basal lamina onion bulb formation, a further increase in Schwann cell density and hypomyelination. Most late onset axonal neuropathy patients showed microangiopathy. In the autopsy case, we observed prominent neuromatous hyperinnervation of the spinal meninges. In four of the six muscle biopsies, we found marked structural mitochondrial abnormalities. These results show that MPZ alterations not only affect myelinated nerve fibers, leading to either primarily demyelinating or axonal changes, but also affect non-myelinated nerve fibers. The autopsy case offers insight into spinal nerve root pathology in MPZ neuropathy. Finally, our data suggest a peculiar association of MPZ mutations with mitochondrial alterations in muscle.
Collapse
Affiliation(s)
- Juliane Bremer
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | - Axel Meinhardt
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | - Istvan Katona
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | - Jan Senderek
- Friedrich Baur Institute at the Department of NeurologyUniversity Hospital, LMU MunichMunichGermany
| | | | - Andreas Roos
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
- Department of NeuropaediatricsUniversity of EssenEssenGermany
| | | | | | - Stefan Nikolin
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | - Kay Nolte
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | - Bernd Sellhaus
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| | | | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin BerlinCampus Virchow‐Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | | | - Eva Neuen‐Jacob
- Department of NeuropathologyUniversity Hospital, Heinrich‐Heine University DüsseldorfDüsseldorfGermany
| | - Chantal Ceuterick de Groote
- Laboratory of Neuromuscular Pathology, Institute Born‐Bunge, and Translational Neurosciences, Faculty of MedicineUniversity of AntwerpBelgium
| | - Peter de Jonghe
- Laboratory of Neuromuscular Pathology, Institute Born‐Bunge, and Translational Neurosciences, Faculty of MedicineUniversity of AntwerpBelgium
- Department of NeurologyUniversity Hospital AntwerpAntwerpBelgium
| | - Vincent Timmerman
- Laboratory of Neuromuscular Pathology, Institute Born‐Bunge, and Translational Neurosciences, Faculty of MedicineUniversity of AntwerpBelgium
- Peripheral Neuropathy Research Group, Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Jonathan Baets
- Laboratory of Neuromuscular Pathology, Institute Born‐Bunge, and Translational Neurosciences, Faculty of MedicineUniversity of AntwerpBelgium
- Department of NeurologyUniversity Hospital AntwerpAntwerpBelgium
| | - Joachim Weis
- Institute of NeuropathologyRWTH Aachen University HospitalAachenGermany
| |
Collapse
|
10
|
Pipis M, Won S, Poh R, Efthymiou S, Polke JM, Skorupinska M, Blake J, Rossor AM, Moran JJ, Munot P, Muntoni F, Laura M, Svaren J, Reilly MM. Post-transcriptional microRNA repression of PMP22 dose in severe Charcot-Marie-Tooth disease type 1. Brain 2023; 146:4025-4032. [PMID: 37337674 PMCID: PMC10545524 DOI: 10.1093/brain/awad203] [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: 02/06/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 06/21/2023] Open
Abstract
Copy number variation (CNV) may lead to pathological traits, and Charcot-Marie-Tooth disease type 1A (CMT1A), the commonest inherited peripheral neuropathy, is due to a genomic duplication encompassing the dosage-sensitive PMP22 gene. MicroRNAs act as repressors on post-transcriptional regulation of gene expression and in rodent models of CMT1A, overexpression of one such microRNA (miR-29a) has been shown to reduce the PMP22 transcript and protein level. Here we present genomic and functional evidence, for the first time in a human CNV-associated phenotype, of the 3' untranslated region (3'-UTR)-mediated role of microRNA repression on gene expression. The proband of the family presented with an early-onset, severe sensorimotor demyelinating neuropathy and harboured a novel de novo deletion in the PMP22 3'-UTR. The deletion is predicted to include the miR-29a seed binding site and transcript analysis of dermal myelinated nerve fibres using a novel platform, revealed a marked increase in PMP22 transcript levels. Functional evidence from Schwann cell lines harbouring the wild-type and mutant 3'-UTR showed significantly increased reporter assay activity in the latter, which was not ameliorated by overexpression of a miR-29a mimic. This shows the importance of miR-29a in regulating PMP22 expression and opens an avenue for therapeutic drug development.
Collapse
Affiliation(s)
- Menelaos Pipis
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Seongsik Won
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, USA
| | - Roy Poh
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Stephanie Efthymiou
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - James M Polke
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Mariola Skorupinska
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Julian Blake
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Department of Clinical Neurophysiology, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
| | - Alexander M Rossor
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - John J Moran
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, USA
| | - Pinki Munot
- Dubowitz Neuromuscular Centre, NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London WC1N 1EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London WC1N 1EH, UK
| | - Matilde Laura
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, USA
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
11
|
Van Lent J, Vendredy L, Adriaenssens E, Da Silva Authier T, Asselbergh B, Kaji M, Weckhuysen S, Van Den Bosch L, Baets J, Timmerman V. Downregulation of PMP22 ameliorates myelin defects in iPSC-derived human organoid cultures of CMT1A. Brain 2023; 146:2885-2896. [PMID: 36511878 PMCID: PMC10316758 DOI: 10.1093/brain/awac475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 10/11/2023] Open
Abstract
Charcot-Marie-Tooth disease is the most common inherited disorder of the PNS. CMT1A accounts for 40-50% of all cases and is caused by a duplication of the PMP22 gene on chromosome 17, leading to dysmyelination in the PNS. Patient-derived models to study such myelination defects are lacking as the in vitro generation of human myelinating Schwann cells has proved to be particularly challenging. Here, we present an induced pluripotent stem cell-derived organoid culture, containing various cell types of the PNS, including myelinating human Schwann cells, which mimics the human PNS. Single-cell analysis confirmed the PNS-like cellular composition and provides insight into the developmental trajectory. We used this organoid model to study disease signatures of CMT1A, revealing early ultrastructural myelin alterations, including increased myelin periodic line distance and hypermyelination of small axons. Furthermore, we observed the presence of onion-bulb-like formations in a later developmental stage. These hallmarks were not present in the CMT1A-corrected isogenic line or in a CMT2A iPSC line, supporting the notion that these alterations are specific to CMT1A. Downregulation of PMP22 expression using short-hairpin RNAs or a combinatorial drug consisting of baclofen, naltrexone hydrochloride and D-sorbitol was able to ameliorate the myelin defects in CMT1A-organoids. In summary, this self-organizing organoid model can capture biologically meaningful features of the disease and capture the physiological complexity, forms an excellent model for studying demyelinating diseases and supports the therapeutic approach of reducing PMP22 expression.
Collapse
Affiliation(s)
- Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
| | - Leen Vendredy
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
| | - Elias Adriaenssens
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
| | - Tatiana Da Silva Authier
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
| | - Bob Asselbergh
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Neuromics Support Facility, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Marcus Kaji
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, University of Antwerp, Antwerp 2610, Belgium
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, University of Antwerp, Antwerp 2610, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp 2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp 2610, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, KU Leuven—University of Leuven, Leuven 3000, Belgium
- VIB-Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven 3000, Belgium
| | - Jonathan Baets
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
- Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp 2610, Belgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, and Translational Neurosciences, Faculty of Medicine, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
12
|
Michailidou I, Vreijling J, Rumpf M, Loos M, Koopmans B, Vlek N, Straat N, Agaser C, Kuipers TB, Mei H, Baas F, Fluiter K. The systemic inhibition of the terminal complement system reduces neuroinflammation but does not improve motor function in mouse models of CMT1A with overexpressed PMP22. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 4:100077. [PMID: 36926597 PMCID: PMC10011818 DOI: 10.1016/j.crneur.2023.100077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 12/13/2022] [Accepted: 01/27/2023] [Indexed: 02/07/2023] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is the most prevalent hereditary demyelinating neuropathy. This autosomal, dominantly inherited disease is caused by a duplication on chromosome 17p which includes the peripheral myelin protein 22 (PMP22) gene. There is clinical evidence that the disability in CMT1A is to a large extend due to axonal damage rather than demyelination. Over-expression of PMP22 is recently thought to impede cholesterol trafficking causing a total shutdown of local cholesterol and lipid synthesis in the Schwann cells, thus disturbing their ability to remyelinate. But there is a large variety in disease burden between CMT1A patients with the same genetic defect, indicating the presence of modifying factors that affect disease severity. One of these potential factors is the immune system. Several reports have described patients with co-occurrence of CMT1A with chronic inflammatory demyelinating disease or Guillain-Barré syndrome. We have previously shown in multiple animal models that the innate immune system and specifically the terminal complement system is a driver of inflammatory demyelination. To test the contribution of the terminal complement system to neuroinflammation and disease progression in CMT1A, we inhibited systemic complement C6 in two transgenic mouse models for CMT1A, the C3-PMP22 and C3-PMP22 c-JunP0Cre models. Both models over-express human PMP22, and one (C3-PMP22 c-JunP0Cre) also has a Schwann cell-specific knockout of c-Jun, a crucial regulator of myelination controlling autophagy. We found that systemic inhibition of C6 using antisense oligonucleotides affects the neuroinflammation, Rho GTPase and ERK/MAPK signalling pathways in the CMT1A mouse models. The cholesterol synthesis pathway remained unaffected. Analysis of motor function during treatment with C6 antisense oligonucleotides did not reveal any significant improvement in the CMT1A mouse models. This study shows that the contribution of the terminal complement system to progressive loss of motor function in the CMT1A mouse models tested is limited.
Collapse
Affiliation(s)
- Iliana Michailidou
- Dept of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Jeroen Vreijling
- Dept of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Matthijs Rumpf
- Dept of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Maarten Loos
- Sylics (Synaptologics B.V.), Bilthoven, the Netherlands
| | | | - Nina Vlek
- Sylics (Synaptologics B.V.), Bilthoven, the Netherlands
| | - Nina Straat
- Sylics (Synaptologics B.V.), Bilthoven, the Netherlands
| | - Cedrick Agaser
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Thomas B Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Frank Baas
- Dept of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Kees Fluiter
- Dept of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
13
|
Clinical findings and genetic analysis of patients with copy number variants involving 17p13.3 using a single nucleotide polymorphism array: a single-center experience. BMC Med Genomics 2022; 15:268. [PMID: 36544138 PMCID: PMC9773569 DOI: 10.1186/s12920-022-01423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND 17p13.3 microdeletions or microduplications (collectively known as copy number variants or CNVs) have been described in individuals with neurodevelopmental disorders. However, 17p13.3 CNVs were rarely reported in fetuses. This study aims to investigate the clinical significance of 17p13.3 CNVs with varied sizes and gene content in prenatal and postnatal samples. METHODS Eight cases with 17p13.3 CNVs out of 8806 samples that had been subjected to single nucleotide polymorphism array analysis were retrospectively analyzed, along with karyotyping, clinical features, and follow-up. RESULTS Eight cases with 17p13.3 CNVs consisted of five fetuses, one aborted embryo and two probands manifested severe congenital defects. The indications of prenatal testing varied considerably for the five fetuses, including ultrasound abnormalities (n = 3), segmental deletions indicated by non-invasive prenatal testing (n = 1), and intellectual disability in the mother of one fetus (n = 1). Of them, two and six harbored copy number gains and losses involving 17p13.3, respectively. The size of the detected 17p13.3 CNVs ranged from 576 kb to 5.7 Mb. Case 1 was diagnosed with 17p13.3 duplication syndrome, and cases 4, 6, and 7 with Miller-Dieker syndrome (MDS). Microdeletions of the 17p13.3 region in two cases (cases 5 and 8) involving YWHAE and CRK, sparing PAFAH1B1, were classified as pathogenic. Case 2 harbored a 576 kb microduplication, encompassing YWHAE and CRK but not PAFAH1B1, which was of maternal origin and considered a variant of uncertain significance. Case 3 carried one 74.2 Mb mosaic duplication of approximately 3.5 on chromosome 17p13.2q25.3, and two deletions at 17p13.3p13.2 and 17q25.3. The karyotype of case 3 was 46,XY,r(17)(p13q25). For five fetuses, only case 2 continued gestation and showed normal development at the age of 15 months; the others were subjected to termination of pregnancy. CONCLUSION The clinical findings of 17p13.3 microdeletions or microduplications varied among subjects, and 17p13.3 CNVs often differ in size and gene content. Microdeletions or microduplications containing the typical MDS region, as well as the microdeletions involving YWHAE and CRK, could be classified as pathogenic. The clinical significance of small duplications including YWHAE and CRK but not PAFAH1B1 remains uncertain, for which parental testing and clinical heterogeneity should be considered in genetic counseling.
Collapse
|
14
|
Roth AR, Li J, Dortch RD. Candidate imaging biomarkers for PMP22-related inherited neuropathies. Ann Clin Transl Neurol 2022; 9:925-935. [PMID: 35656877 PMCID: PMC9268861 DOI: 10.1002/acn3.51561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Charcot-Marie-Tooth type 1A (CMT1A) and hereditary neuropathy with liability to pressure palsy (HNPP) are caused by mutations to the peripheral myelin protein 22 (PMP22) gene. A need exists for sensitive and reliable biomarkers of progression and treatment response. Magnetic resonance imaging (MRI) metrics of nerve pathology and morphology were investigated for this purpose. METHODS MRI was performed at 3.0 T in the thigh of CMT1A (N = 11) and HNPP patients (N = 12) and controls (N = 23). Three potential imaging biomarkers of the sciatic nerve were investigated: 1) magnetization transfer ratio (MTR), which assays myelin content, and 2) cross-sectional area (CSA) and 3) circularity, which assay morphological changes. Potential imaging biomarkers were compared across cohorts and assessed for relationships with disability in the legs (CMTESL ), compound motor action potentials (CMAP), and motor conduction velocities (MCV). Inter-rater reliability and test-retest repeatability were established for each imaging metric. RESULTS Significant differences in MTR, CSA, and circularity were observed in CMT1A relative to controls (p = 0.02, p < 0.001, and p = 0.003, respectively, via Wilcoxon rank-sum tests). Differences were not observed in the HNPP cohort. Significant relationships were observed between MTR and clinical metrics (CMTESL : p = 0.003, CMAP: p = 0.03, MCV: p = 0.01); and between CSA and electrophysiology (CMAP: p = 0.002, MCV: p < 0.001). All metrics were reliable and repeatable with MTR the most reliable (intraclass correlation coefficient [ICC] >0.999, CV = 0.30%) and repeatable (ICC = 0.84, CV = 3.16%). INTERPRETATION MTR, CSA, and circularity showed promise as reliable and sensitive biomarkers of CMT1A, but not HNPP. These warrant longitudinal investigation as response biomarkers in upcoming clinical trials of CMT1A, while other methods should be considered for HNPP.
Collapse
Affiliation(s)
- Alison R. Roth
- Division of Neuroimaging ResearchBarrow Neurological InstitutePhoenixArizonaUSA
| | - Jun Li
- Department of NeurologyVanderbilt UniversityNashvilleTennesseeUSA
- Department of NeurologyWayne State UniversityDetroitMichiganUSA
| | - Richard D. Dortch
- Division of Neuroimaging ResearchBarrow Neurological InstitutePhoenixArizonaUSA
- Vanderbilt University Institute of Imaging ScienceVanderbilt UniversityNashvilleTennesseeUSA
- Department of Radiology and Radiological SciencesVanderbilt UniversityNashvilleTennesseeUSA
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| |
Collapse
|
15
|
Yalcouyé A, Esoh K, Guida L, Wonkam A. Current profile of Charcot-Marie-Tooth disease in Africa: A systematic review. J Peripher Nerv Syst 2022; 27:100-112. [PMID: 35383421 PMCID: PMC9322329 DOI: 10.1111/jns.12489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/17/2022] [Accepted: 02/25/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Charcot-Marie-Tooth disease (CMT) is the most common inherited peripheral neuropathy characterised by a high clinical and genetic heterogeneity. While most cases were described in populations with Caucasian ancestry, genetic research on CMT in Africa is scant. Only a few cases of CMT have been reported, mainly from North Africa. The current study aimed to summarise available data on CMT in Africa, with emphasis on the epidemiological, clinical, and genetic features. METHODS We searched PubMed, Scopus, Web of Sciences, and the African Journal Online for articles published from the database inception until April 2021 using specific keywords. A total of 398 articles were screened, and 28 fulfilled our selection criteria. RESULTS A total of 107 families totalling 185 patients were reported. Most studies were reported from North Africa (n = 22). The demyelinating form of CMT was the commonest subtype, and the phenotype varied greatly between families, and one family (1%) of CMT associated with hearing impairment was reported. The inheritance pattern was autosomal recessive in 91.2% (n = 97/107) of families. CMT-associated variants were reported in 11 genes: LMNA, GDAP1, GJB1, MPZ, MTMR13, MTMR2, PRX, FGD4/FRABIN, PMP22, SH3TC2, and GARS. The most common genes reported are LMNA, GDAP1, and SH3TC2 and have been found mostly in Northern African populations. INTERPRETATION This study reveals that CMT is not rare in Africa, and describes the current clinical and genetic profile. The review emphasised the urgent need to invest in genetic research to inform counselling, prevention, and care for CMT in numerous settings on the continent.
Collapse
Affiliation(s)
- Abdoulaye Yalcouyé
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali.,Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Kevin Esoh
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Landouré Guida
- Faculté de Médecine et d'Odontostomatologie, USTTB, Bamako, Mali.,Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA.,Service de Neurologie, Centre Hospitalier Universitaire du Point "G", Bamako, Mali
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,McKusick-Nathans Institute, and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Winter N, Vittore D, Gess B, Schulz JB, Grimm A, Dohrn MF. New Keys to Early Diagnosis: Muscle Echogenicity, Nerve Ultrasound Patterns, Electrodiagnostic, and Clinical Parameters in 150 Patients with Hereditary Polyneuropathies. Neurotherapeutics 2021; 18:2425-2435. [PMID: 34708324 PMCID: PMC8804010 DOI: 10.1007/s13311-021-01141-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2021] [Indexed: 11/26/2022] Open
Abstract
Hereditary neuropathies are of variable genotype and phenotype. With upcoming therapies, there is urgent need for early disease recognition and outcome measures. High-resolution nerve and muscle ultrasound is a dynamic, non-invasive, well-established tool in the field of inflammatory and traumatic neuropathies. In this study, we defined nerve and muscle ultrasound parameters as recognition and progression markers in 150 patients with genetically confirmed hereditary neuropathies, including Charcot-Marie-Tooth (CMT) disease (CMT1A, n = 55; other CMT1/4, n = 28; axonal CMT, n = 15; CMTX, n = 15), hereditary neuropathy with liability to pressure palsies (HNPP, n = 16), hereditary transthyretin-amyloidosis (ATTRv, n = 14), and Fabry's disease (n = 7). The CMT1A, followed by the CMT1/4 group, had the most homogeneous enlargement of the nerve cross-sectional areas (CSA) in the ultrasound pattern sum (UPSS) and homogeneity score. Entrapment scores were highest in HNPP, ATTRv amyloidosis, and Fabry's disease patients. In demyelinating neuropathies, the CSA correlated inversely with nerve conduction studies. The muscle echo intensity was significantly highest in the clinically most affected muscles, which was independent from the underlying disease cause and correlated with muscle strength and disease duration. Further correlations were seen with combined clinical (CMTES-2) and electrophysiological (CMTNS-2) scores of disease severity. We conclude that nerve ultrasound is a helpful tool to distinguish different types of hereditary neuropathies by pattern recognition, whereas muscle ultrasound is an objective parameter for disease severity. The implementation of neuromuscular ultrasound might enrich diagnostic procedures both in clinical routines and research.
Collapse
Affiliation(s)
- Natalie Winter
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Debora Vittore
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Burkhard Gess
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jörg B Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Jülich Aachen Research Alliance (JARA), FZ Jülich and RWTH University, Jülich, Germany
| | - Alexander Grimm
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.
| | - Maike F Dohrn
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Bosco L, Falzone YM, Previtali SC. Animal Models as a Tool to Design Therapeutical Strategies for CMT-like Hereditary Neuropathies. Brain Sci 2021; 11:1237. [PMID: 34573256 PMCID: PMC8465478 DOI: 10.3390/brainsci11091237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Since ancient times, animal models have provided fundamental information in medical knowledge. This also applies for discoveries in the field of inherited peripheral neuropathies (IPNs), where they have been instrumental for our understanding of nerve development, pathogenesis of neuropathy, molecules and pathways involved and to design potential therapies. In this review, we briefly describe how animal models have been used in ancient medicine until the use of rodents as the prevalent model in present times. We then travel along different examples of how rodents have been used to improve our understanding of IPNs. We do not intend to describe all discoveries and animal models developed for IPNs, but just to touch on a few arbitrary and paradigmatic examples, taken from our direct experience or from literature. The idea is to show how strategies have been developed to finally arrive to possible treatments for IPNs.
Collapse
Affiliation(s)
| | | | - Stefano Carlo Previtali
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.B.); (Y.M.F.)
| |
Collapse
|
18
|
Uchôa Cavalcanti EB, Santos SCDL, Martins CES, de Carvalho DR, Rizzo IMPDO, Freitas MCDNB, da Silva Freitas D, de Souza FS, Junior AM, do Nascimento OJM. Charcot-Marie-Tooth disease: Genetic profile of patients from a large Brazilian neuromuscular reference center. J Peripher Nerv Syst 2021; 26:290-297. [PMID: 34190362 DOI: 10.1111/jns.12458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 11/28/2022]
Abstract
This study aimed to describe the clinical, genetic, and epidemiological features of Charcot-Marie-Tooth disease (CMT) in Brazilian patients from a tertiary center, and to compare our data with previously published findings. This retrospective observational study conducted between February 2015 and July 2020 evaluated 503 patients (94 families and 192 unrelated individuals), diagnosed with CMT. Clinical and neurophysiological data were obtained from electronic medical records and blood samples were used for genetic analyses. Multiplex ligation-dependent probe amplification was used to assess duplications/deletions in PMP22. Sanger sequencing of GJB1 was performed in cases of suspected demyelinating CMT. Targeted gene panel sequencing was used for the remaining negative demyelinating cases and all axonal CMT cases. The first decade of life was the most common period of disease onset. In all, 353 patients had demyelinating CMT, 39 had intermediate CMT, and 111 had axonal CMT. Pathogenic or likely pathogenic variants were identified in 197 index cases. The most common causative genes among probands were PMP22 (duplication) (n = 116, 58.88%), GJB1 (n = 23, 11.67%), MFN2 (n = 12, 6.09%), GDAP1 (n = 7, 3.55%), MPZ (n = 6, 3.05%), PMP22 (point mutation) (n = 6, 3.05%), NEFL (n = 3, 1.52%), SBF2 (n = 3, 1.52%), and SH3TC2 (n = 3, 1.52%). Other identified variants were ≤1% of index cases. This study provides further data on the frequency of CMT subtypes in a Brazilian clinical-based population and highlights the importance of rarer and previously undiagnosed variants in clinical practice.
Collapse
|
19
|
Miniou P, Fontes M. Therapeutic Development in Charcot Marie Tooth Type 1 Disease. Int J Mol Sci 2021; 22:ijms22136755. [PMID: 34201736 PMCID: PMC8268813 DOI: 10.3390/ijms22136755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 01/04/2023] Open
Abstract
Charcot–Marie–Tooth disease (CMT) is the most frequent hereditary peripheral neuropathies. It is subdivided in two main groups, demyelinating (CMT1) and axonal (CMT2). CMT1 forms are the most frequent. The goal of this review is to present published data on 1—cellular and animal models having opened new potential therapeutic approaches. 2—exploration of these tracks, including clinical trials. The first conclusion is the great increase of publications on CMT1 subtypes since 2000. We discussed two points that should be considered in the therapeutic development toward a regulatory-approved therapy to be proposed to patients. The first point concerns long term safety if treatments will be a long-term process. The second point relates to the evaluation of treatment efficiency. Degradation of CMT clinical phenotype is not linear and progressive.
Collapse
Affiliation(s)
- Pierre Miniou
- InFlectis BioScience SAS, 21 Rue La Noue Bras de Fer, 44200 Nantes, France;
| | - Michel Fontes
- Centre de recherche en CardioVasculaire et Nutrition, Aix-Marseille Université, INRA 1260—INSERM 1263, 13005 Marseille, France
- Repositioning SAS, 8 Rue Napoleon, 20210 Calenzana, France
- Correspondence:
| |
Collapse
|
20
|
Wang H, Davison M, Wang K, Xia TH, Call KM, Luo J, Wu X, Zuccarino R, Bacha A, Bai Y, Gutmann L, Feely SME, Grider T, Rossor AM, Reilly MM, Shy ME, Svaren J. MicroRNAs as Biomarkers of Charcot-Marie-Tooth Disease Type 1A. Neurology 2021; 97:e489-e500. [PMID: 34031204 DOI: 10.1212/wnl.0000000000012266] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To determine whether microRNAs (miRs) are elevated in the plasma of individuals with the inherited peripheral neuropathy Charcot-Marie-Tooth disease type 1A (CMT1A), miR profiling was employed to compare control and CMT1A plasma. METHODS We performed a screen of CMT1A and control plasma samples to identify miRs that are elevated in CMT1A using next-generation sequencing, followed by validation of selected miRs by quantitative PCR, and correlation with protein biomarkers and clinical data: Rasch-modified CMT Examination and Neuropathy Scores, ulnar compound muscle action potentials, and motor nerve conduction velocities. RESULTS After an initial pilot screen, a broader screen confirmed elevated levels of several muscle-associated miRNAs (miR1, -133a, -133b, and -206, known as myomiRs) along with a set of miRs that are highly expressed in Schwann cells of peripheral nerve. Comparison to other candidate biomarkers for CMT1A (e.g., neurofilament light) measured on the same sample set shows a comparable elevation of several miRs (e.g., miR133a, -206, -223) and ability to discriminate cases from controls. Neurofilament light levels were most highly correlated with miR133a. In addition, the putative Schwann cell miRs (e.g., miR223, -199a, -328, -409, -431) correlate with the recently described transmembrane protease serine 5 (TMPRSS5) protein biomarker that is most highly expressed in Schwann cells and also elevated in CMT1A plasma. CONCLUSIONS These studies identify a set of miRs that are candidate biomarkers for clinical trials in CMT1A. Some of the miRs may reflect Schwann cell processes that underlie the pathogenesis of the disease. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that a set of plasma miRs are elevated in patients with CMT1A.
Collapse
Affiliation(s)
- Hongge Wang
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Matthew Davison
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Kathryn Wang
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Tai-He Xia
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Katherine M Call
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Jun Luo
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Xingyao Wu
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Riccardo Zuccarino
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Alexa Bacha
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Yunhong Bai
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Laurie Gutmann
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Shawna M E Feely
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Tiffany Grider
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Alexander M Rossor
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Mary M Reilly
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Michael E Shy
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - John Svaren
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison.
| |
Collapse
|
21
|
Jennings MJ, Lochmüller A, Atalaia A, Horvath R. Targeted Therapies for Hereditary Peripheral Neuropathies: Systematic Review and Steps Towards a 'treatabolome'. J Neuromuscul Dis 2021; 8:383-400. [PMID: 32773395 PMCID: PMC8203235 DOI: 10.3233/jnd-200546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Hereditary peripheral neuropathies are inherited disorders affecting the peripheral nervous system, including Charcot-Marie-Tooth disease, familial amyloid polyneuropathy and hereditary sensory and motor neuropathies. While the molecular basis of hereditary peripheral neuropathies has been extensively researched, interventional trials of pharmacological therapies are lacking. Objective: We collated evidence for the effectiveness of pharmacological and gene-based treatments for hereditary peripheral neuropathies. Methods: We searched several databases for randomised controlled trials (RCT), observational studies and case reports of therapies in hereditary peripheral neuropathies. Two investigators extracted and analysed the data independently, assessing study quality using the Oxford Centre for Evidence Based Medicine 2011 Levels of Evidence in conjunction with the Jadad scale. Results: Of the 2046 studies initially identified, 119 trials met our inclusion criteria, of which only 34 were carried over into our final analysis. Ascorbic acid was shown to have no therapeutic benefit in CMT1A, while a combination of baclofen, naltrexone and sorbitol (PXT3003) demonstrated some efficacy, but phase III data are incomplete. In TTR-related amyloid polyneuropathy tafamidis, patisiran, inotersen and revusiran showed significant benefit in high quality RCTs. Smaller studies showed the efficacy of L-serine for SPTLC1-related hereditary sensory neuropathy, riboflavin for Brown-Vialetto-Van Laere syndrome (SLC52A2/3) and phytanic acid-poor diet in Refsum disease (PHYH). Conclusions: The ‘treatable’ variants highlighted in this project will be flagged in the treatabolome database to alert clinicians at the time of the diagnosis and enable timely treatment of patients with hereditary peripheral neuropathies.
Collapse
Affiliation(s)
- Matthew J Jennings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Antonio Atalaia
- Center of Research in Myology, Sorbonne Université - Inserm UMRS 974, Institut de Myologie, G.H. Pitie-Salpetriere, Paris, France
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Han L, Huang Y, Nie Y, Li J, Chen G, Tu S, Shen P, Chen C. A novel PMP22 insertion mutation causing Charcot-Marie-Tooth disease type 3: A case report. Medicine (Baltimore) 2021; 100:e25163. [PMID: 33726003 PMCID: PMC7982204 DOI: 10.1097/md.0000000000025163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/25/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Charcot-Marie-Tooth disease (CMT) is a group of hereditary neuropathies with clinical features of muscle atrophy, sensory loss, and foot deformities. CMT is related to a number of genes, such as peripheral myelin protein 22 gene (PMP22). Missense mutations, small deletion mutations, and duplications of PMP22 are common in CMT patients, but few insertion mutation cases of PMP22 have been reported. PATIENT CONCERNS A 26-year-old male patient with the complaint of general weakness, peroneal atrophy, and deformities in the extremities visited our hospital. The patient was born with bilateral thumbs and feet dystonia. Additionally, delayed feet arch development and delayed walking was observed when he was a child. DIAGNOSIS Using whole-exome sequencing and electrophysiological test, we identified a novel insertion mutation of PMP22 (NM_153322, c.54_55insGTGCTG, p.(L19delinsVLL)) in a 26-year-old male patient with peroneal atrophy and nerve conduction was not elicited in electromyography (EMG) study. The Protein Variation Effect Analyzer (PROVEAN) program analysis predicted that the variant is likely to be "deleterious." SWISS-MODEL program predicted that alpha helix in original location was disrupted by inserted 6 bases, which may account for the occurrence of CMT3. INTERVENTIONS The patient received symptomatic and supportive treatments, and routine rehabilitation exercises during hospitalization. OUTCOMES The condition of the patient was improved, but the disease could not be cured. At 1- and 3-months follow-up, manifestations of the patient were unchanged, and he could take care of himself. LESSONS Our findings link a novel PMP22 mutation with a clinical diagnosis of CMT3. The link between gene variation and CMT phenotype may help to reveal the structure and function of PMP22 protein and the pathogenesis of CMT. This study adds further support to the heterogeneity of PMP22 related CMT and provides solid functional evidence for the pathogenicity of the p.(L19delinsVLL) PMP22 variant. Moreover, with the development of high-throughput sequencing technology, the combination of next-generation sequencing (NGS) and conventional Sanger sequencing is becoming one of the comprehensive, inexpensive, and convenient tools for genetic diagnosis of CMT.
Collapse
Affiliation(s)
- Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Yanjing Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Yuan Nie
- Rehabilitation Center, Qijiang District Hospital of Traditional Chinese Medicine, 50 Dashi Road of Wenlong Avenue, Chongqing
| | - Jing Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Gang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Pan Shen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan
| | - Chao Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| |
Collapse
|
23
|
Safka Brozkova D, Stojkovic T, Haberlová J, Mazanec R, Windhager R, Fernandes Rosenegger P, Hacker S, Züchner S, Kochański A, Leonard-Louis S, Francou B, Latour P, Senderek J, Seeman P, Auer-Grumbach M. Demyelinating Charcot-Marie-Tooth neuropathy associated with FBLN5 mutations. Eur J Neurol 2020; 27:2568-2574. [PMID: 32757322 DOI: 10.1111/ene.14463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/29/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Charcot-Marie-Tooth disease type 1 (CMT1) is a group of autosomal dominantly inherited demyelinating sensorimotor neuropathies. Symptoms usually start in the first to second decade and include distal muscle weakness and wasting, sensory disturbances and foot deformities. The most frequent cause is a duplication of PMP22 whilst point mutations in PMP22 and other genes are rare causes. Recently, FBLN5 mutations have been reported in CMT1 families. METHODS Individuals with FBLN5-associated CMT1 were compiled from clinical and research genetic testing laboratories. Clinical data were extracted from medical records or obtained during patients' visits at our centres or primary care sites. RESULTS Nineteen CMT1 families containing 38 carriers of three different FBLN5 missense variants were identified and a mutational hotspot at c.1117C>T (p.Arg373Cys) was confirmed. Compared to patients with the common PMP22 duplication, individuals with FBLN5 variants had a later age of diagnosis (third to fifth decade) and less severely reduced motor median nerve conduction velocities (around 31 m/s). The most frequent clinical presentations were prominent sensory disturbances and painful sensations, often as initial symptom and pronounced in the upper limbs, contrasting with rather mild to moderate motor deficits. CONCLUSIONS Our study confirms the relevance of FBLN5 mutations in CMT1. It is proposed to include FBLN5 in the genetic work-up of individuals suspected with CMT1, particularly when diagnosis is established beyond the first and second decade and comparably moderate motor deficits contrast with early and marked sensory involvement. FBLN5-associated CMT1 has a recognizable clinical phenotype and should be referred to as CMT1H according to the current classification scheme.
Collapse
Affiliation(s)
- D Safka Brozkova
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - T Stojkovic
- Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Institut de Myologie, APHP, G-H Pitié-Salpêtrière, Paris, France
| | - J Haberlová
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - R Mazanec
- Department of Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - R Windhager
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - P Fernandes Rosenegger
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - S Hacker
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - S Züchner
- Dr John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Kochański
- Neuromuscular Unit, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - S Leonard-Louis
- Unité de Pathologie Neuromusculaire, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, APHP, G-H Pitié-Salpêtrière, Paris, France
| | - B Francou
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, APHP, Hôpital Kremlin-Bicêtre, Paris, France
| | - P Latour
- Service de Biochimie et Biologie Moléculaire Grand Est, CHU de Lyon, GH Est, Bron, France
| | - J Senderek
- Department of Neurology, Friedrich-Baur-Institute, LMU Munich, Munich, Germany
| | - P Seeman
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - M Auer-Grumbach
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Caillaud M, Msheik Z, Ndong-Ntoutoume GMA, Vignaud L, Richard L, Favreau F, Faye PA, Sturtz F, Granet R, Vallat JM, Sol V, Desmoulière A, Billet F. Curcumin-cyclodextrin/cellulose nanocrystals improve the phenotype of Charcot-Marie-Tooth-1A transgenic rats through the reduction of oxidative stress. Free Radic Biol Med 2020; 161:246-262. [PMID: 32980538 DOI: 10.1016/j.freeradbiomed.2020.09.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/09/2020] [Accepted: 09/20/2020] [Indexed: 12/16/2022]
Abstract
The most prevalent form of Charcot-Marie-Tooth disease (CMT type 1A) is characterized by duplication of the PMP22 gene, peripheral dysmyelination and decreased nerve conduction velocities leading to muscle weakness. Recently, oxidative stress was reported as a feature in CMT1A patients. Curcumin exhibits antioxidant activities and has shown beneficial properties on peripheral nerves. However, curcumin presents unfavorable pharmacokinetics. We developed curcumin-cyclodextrin/cellulose nanocrystals (Nano-Cur) to bypass this limitation. The present study investigated the therapeutic potential of Nano-Cur in vitro in Schwann cells (SCs) and in vivo in the transgenic CMT1A rat model. In vitro, Nano-Cur treatment (0.01 μM for 8 h) reduced reactive oxygen species and improved mitochondrial membrane potential in CMT1A SCs. Moreover, Nano-Cur treatment (0.01 μM for 1 week) increased the expression of myelin basic protein in SC/neuron co-cultures. Preliminary in vivo experiments carried out in WT rats showed that intraperitoneal (i.p.) injection of Nano-Cur treatment containing 0.2 mg/kg of curcumin strongly enhanced the bioavailability of curcumin. Afterwards, in 1-month-old male CMT1A rats, Nano-Cur treatment (0.2 mg/kg/day, i.p. for 8 weeks) significantly improved sensori-motor functions (grip strength, balance performance, and mechanical and thermal sensitivities). Importantly, sensory and motor nerve conduction velocities were improved. Further histological and biochemical analyses indicated that myelin sheath thickness and myelin protein expression (myelin protein zero and PMP22) were increased. In addition, oxidative stress markers were decreased in the sciatic nerve and gastrocnemius muscle. Finally, Nrf2 expression and some major antioxidant enzymes were increased in sciatic nerve. Therefore, Nano-Cur significantly improved cellular, electrophysiological, and functional features of CMT1A rats.
Collapse
Affiliation(s)
- Martial Caillaud
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France; Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Zeina Msheik
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France
| | - Gautier M-A Ndong-Ntoutoume
- EA7500, PEIRENE Laboratory, Faculty of Science and Technology, University of Limoges, F-87000, Limoges, France
| | - Laetitia Vignaud
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France
| | - Laurence Richard
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France; Reference Center for Rare Peripheral Neuropathies, Department of Neurology, University Hospital of Limoges, F-87000, Limoges, France
| | - Frédéric Favreau
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France; Department of Biochemistry, University Hospital of Limoges, F-87000, Limoges, France
| | - Pierre-Antoine Faye
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France; Department of Biochemistry, University Hospital of Limoges, F-87000, Limoges, France
| | - Franck Sturtz
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France; Department of Biochemistry, University Hospital of Limoges, F-87000, Limoges, France
| | - Robert Granet
- EA7500, PEIRENE Laboratory, Faculty of Science and Technology, University of Limoges, F-87000, Limoges, France
| | - Jean-Michel Vallat
- Reference Center for Rare Peripheral Neuropathies, Department of Neurology, University Hospital of Limoges, F-87000, Limoges, France
| | - Vincent Sol
- EA7500, PEIRENE Laboratory, Faculty of Science and Technology, University of Limoges, F-87000, Limoges, France
| | - Alexis Desmoulière
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France
| | - Fabrice Billet
- EA6309, Myelin Maintenance and Peripheral Neuropathies, University of Limoges, Faculties of Medicine and Pharmacy, F-87000, Limoges, France.
| |
Collapse
|
25
|
Reilly MM, Rossor AM. Humans: the ultimate animal models. J Neurol Neurosurg Psychiatry 2020; 91:1132-1136. [PMID: 32769113 PMCID: PMC7415072 DOI: 10.1136/jnnp-2020-323016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Alexander M Rossor
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
26
|
Lupski JR, Timmerman V. The CMT1A duplication. MED GENET-BERLIN 2020. [DOI: 10.1515/medgen-2020-2030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
27
|
Kurth I. Peripheral Neuropathies. MED GENET-BERLIN 2020. [DOI: 10.1515/medgen-2020-2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Ingo Kurth
- Institute of Human Genetics, Medical Faculty , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
28
|
Pantera H, Hu B, Moiseev D, Dunham C, Rashid J, Moran JJ, Krentz K, Rubinstein CD, Won S, Li J, Svaren J. Pmp22 super-enhancer deletion causes tomacula formation and conduction block in peripheral nerves. Hum Mol Genet 2020; 29:1689-1699. [PMID: 32356557 PMCID: PMC7322568 DOI: 10.1093/hmg/ddaa082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/07/2020] [Accepted: 04/24/2020] [Indexed: 11/12/2022] Open
Abstract
Copy number variation of the peripheral nerve myelin gene Peripheral Myelin Protein 22 (PMP22) causes multiple forms of inherited peripheral neuropathy. The duplication of a 1.4 Mb segment surrounding this gene in chromosome 17p12 (c17p12) causes the most common form of Charcot-Marie-Tooth disease type 1A, whereas the reciprocal deletion of this gene causes a separate neuropathy termed hereditary neuropathy with liability to pressure palsies (HNPP). PMP22 is robustly induced in Schwann cells in early postnatal development, and several transcription factors and their cognate regulatory elements have been implicated in coordinating the gene's proper expression. We previously found that a distal super-enhancer domain was important for Pmp22 expression in vitro, with particular impact on a Schwann cell-specific alternative promoter. Here, we investigate the consequences of deleting this super-enhancer in vivo. We find that loss of the super-enhancer in mice reduces Pmp22 expression throughout development and into adulthood, with greater impact on the Schwann cell-specific promoter. Additionally, these mice display tomacula formed by excessive myelin folding, a pathological hallmark of HNPP, as have been previously observed in heterozygous Pmp22 mice as well as sural biopsies from patients with HNPP. Our findings demonstrate a mechanism by which smaller copy number variations, not including the Pmp22 gene, are sufficient to reduce gene expression and phenocopy a peripheral neuropathy caused by the HNPP-associated deletion encompassing PMP22.
Collapse
Affiliation(s)
- Harrison Pantera
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bo Hu
- Department of Neurology and Translational Neuroscience Initiative, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Daniel Moiseev
- Department of Neurology and Translational Neuroscience Initiative, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Chris Dunham
- Department of Neurology and Translational Neuroscience Initiative, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Jibraan Rashid
- Department of Neurology and Translational Neuroscience Initiative, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - John J Moran
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kathleen Krentz
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - C Dustin Rubinstein
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Seongsik Won
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jun Li
- Department of Neurology and Translational Neuroscience Initiative, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
29
|
Prukop T, Wernick S, Boussicault L, Ewers D, Jäger K, Adam J, Winter L, Quintes S, Linhoff L, Barrantes-Freer A, Bartl M, Czesnik D, Zschüntzsch J, Schmidt J, Primas G, Laffaire J, Rinaudo P, Brureau A, Nabirotchkin S, Schwab MH, Nave KA, Hajj R, Cohen D, Sereda MW. Synergistic PXT3003 therapy uncouples neuromuscular function from dysmyelination in male Charcot-Marie-Tooth disease type 1A (CMT1A) rats. J Neurosci Res 2020; 98:1933-1952. [PMID: 32588471 DOI: 10.1002/jnr.24679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/13/2020] [Accepted: 05/31/2020] [Indexed: 12/11/2022]
Abstract
Charcot-Marie-Tooth disease 1 A (CMT1A) is caused by an intrachromosomal duplication of the gene encoding for PMP22 leading to peripheral nerve dysmyelination, axonal loss, and progressive muscle weakness. No therapy is available. PXT3003 is a low-dose combination of baclofen, naltrexone, and sorbitol which has been shown to improve disease symptoms in Pmp22 transgenic rats, a bona fide model of CMT1A disease. However, the superiority of PXT3003 over its single components or dual combinations have not been tested. Here, we show that in a dorsal root ganglion (DRG) co-culture system derived from transgenic rats, PXT3003 induced myelination when compared to its single and dual components. Applying a clinically relevant ("translational") study design in adult male CMT1A rats for 3 months, PXT3003, but not its dual components, resulted in improved performance in behavioral motor and sensory endpoints when compared to placebo. Unexpectedly, we observed only a marginally increased number of myelinated axons in nerves from PXT3003-treated CMT1A rats. However, in electrophysiology, motor latencies correlated with increased grip strength indicating a possible effect of PXT3003 on neuromuscular junctions (NMJs) and muscle fiber pathology. Indeed, PXT3003-treated CMT1A rats displayed an increased perimeter of individual NMJs and a larger number of functional NMJs. Moreover, muscles of PXT3003 CMT1A rats displayed less neurogenic atrophy and a shift toward fast contracting muscle fibers. We suggest that ameliorated motor function in PXT3003-treated CMT1A rats result from restored NMJ function and muscle innervation, independent from myelination.
Collapse
Affiliation(s)
- Thomas Prukop
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephanie Wernick
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - David Ewers
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Karoline Jäger
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Julia Adam
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Lorenz Winter
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Susanne Quintes
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Lisa Linhoff
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Michael Bartl
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk Czesnik
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | - Markus H Schwab
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | | | | | - Michael W Sereda
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
30
|
Lee JS, Lee JY, Song DW, Bae HS, Doo HM, Yu HS, Lee KJ, Kim HK, Hwang H, Kwak G, Kim D, Kim S, Hong YB, Lee JM, Choi BO. Targeted PMP22 TATA-box editing by CRISPR/Cas9 reduces demyelinating neuropathy of Charcot-Marie-Tooth disease type 1A in mice. Nucleic Acids Res 2020; 48:130-140. [PMID: 31713617 PMCID: PMC7145652 DOI: 10.1093/nar/gkz1070] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 12/26/2022] Open
Abstract
Charcot-Marie-Tooth 1A (CMT1A) is the most common inherited neuropathy without a known therapy, which is caused by a 1.4 Mb duplication on human chromosome 17, which includes the gene encoding the peripheral myelin protein of 22 kDa (PMP22). Overexpressed PMP22 protein from its gene duplication is thought to cause demyelination and subsequently axonal degeneration in the peripheral nervous system (PNS). Here, we targeted TATA-box of human PMP22 promoter to normalize overexpressed PMP22 level in C22 mice, a mouse model of CMT1A harboring multiple copies of human PMP22. Direct local intraneural delivery of CRISPR/Cas9 designed to target TATA-box of PMP22 before the onset of disease, downregulates gene expression of PMP22 and preserves both myelin and axons. Notably, the same approach was effective in partial rescue of demyelination even after the onset of disease. Collectively, our data present a proof-of-concept that CRISPR/Cas9-mediated targeting of TATA-box can be utilized to treat CMT1A.
Collapse
Affiliation(s)
- Ji-Su Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | | | | | | | - Hyun M Doo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Ho S Yu
- ToolGen, Inc., Seoul, 08501, Korea
| | | | - Hee K Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Hyun Hwang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Geon Kwak
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Daesik Kim
- Center for Genome Engineering, Institute for Basic Science (IBS), Seoul, 08826, Korea
- Department of Chemistry, Seoul National University, Seoul, 08826, Korea
| | | | - Young B Hong
- Department of Biochemistry, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Jung M Lee
- School of Life Science, Handong Global University, Pohang 37554, Korea
| | - Byung-Ok Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| |
Collapse
|
31
|
Wang H, Davison M, Wang K, Xia T, Kramer M, Call K, Luo J, Wu X, Zuccarino R, Bacon C, Bai Y, Moran JJ, Gutmann L, Feely SME, Grider T, Rossor AM, Reilly MM, Svaren J, Shy ME. Transmembrane protease serine 5: a novel Schwann cell plasma marker for CMT1A. Ann Clin Transl Neurol 2020; 7:69-82. [PMID: 31833243 PMCID: PMC6952315 DOI: 10.1002/acn3.50965] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/13/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Development of biomarkers for Charcot-Marie-Tooth (CMT) disease is critical for implementing effective clinical trials. The most common form of CMT, type 1A, is caused by a genomic duplication surrounding the PMP22 gene. A recent report (Neurology 2018;90:e518-3524) showed elevation of neurofilament light (NfL) in plasma of CMT1A disease patients, which correlated with disease severity. However, no plasma/serum biomarker has been identified that is specific to Schwann cells, the most directly affected cells in CMT1A. METHODS We used the Olink immuno PCR platform to profile CMT1A patient (n = 47, 2 cohorts) and normal control plasma (n = 41, two cohorts) on five different Olink panels to screen 398 unique proteins. RESULTS The TMPRSS5 protein (Transmembrane protease serine 5) was elevated 2.07-fold (P = <0.0001) in two independent cohorts of CMT1A samples relative to controls. TMPRSS5 is most highly expressed in Schwann cells of peripheral nerve. Consistent with early myelination deficits in CMT1A, TMPRSS5 was not significantly correlated with disease score (CMTES-R, CMTNS-R), nerve conduction velocities (Ulnar CMAP, Ulnar MNCV), or with age. TMPRSS5 was not significantly elevated in smaller sample sets from patients with CMT2A, CMT2E, CMT1B, or CMT1X. The Olink immuno PCR assays confirmed elevated levels of NfL (average 1.58-fold, P < 0.0001), which correlated with CMT1A patient disease score. INTERPRETATION These data identify the first Schwann cell-specific protein that is elevated in plasma of CMT1A patients, and may provide a disease marker and a potentially treatment-responsive biomarker with good disease specificity for clinical trials.
Collapse
Affiliation(s)
- Hongge Wang
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Matthew Davison
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Kathryn Wang
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Tai‐He Xia
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Martin Kramer
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Katherine Call
- Translational SciencesSanofi ResearchSanofiFraminghamMassachusetts
| | - Jun Luo
- Research StatisticsSanofi ResearchSanofiFraminghamMassachusetts
| | - Xingyao Wu
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Riccardo Zuccarino
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Chelsea Bacon
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Yunhong Bai
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - John J. Moran
- Waisman Center and Department of Comparative BiosciencesUniversity of WisconsinMadisonWisconsin
| | - Laurie Gutmann
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Shawna M. E. Feely
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Tiffany Grider
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| | - Alexander M. Rossor
- National Hospital for Neurology and NeurosurgeryUniversity College LondonLondonUnited Kingdom
| | - Mary M. Reilly
- National Hospital for Neurology and NeurosurgeryUniversity College LondonLondonUnited Kingdom
| | - John Svaren
- Waisman Center and Department of Comparative BiosciencesUniversity of WisconsinMadisonWisconsin
| | - Michael E. Shy
- Department of NeurologyCarver College of MedicineUniversity of IowaIowa CityIowa
| |
Collapse
|
32
|
Serfecz J, Bazick H, Al Salihi MO, Turner P, Fields C, Cruz P, Renne R, Notterpek L. Downregulation of the human peripheral myelin protein 22 gene by miR-29a in cellular models of Charcot-Marie-Tooth disease. Gene Ther 2019; 26:455-464. [PMID: 31455873 PMCID: PMC6920087 DOI: 10.1038/s41434-019-0098-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022]
Abstract
The majority of hereditary neuropathies are caused by duplication of the peripheral myelin protein 22 (PMP22) gene. Therefore, mechanisms to suppress the expression of the PMP22 gene have high therapeutic significance. Here we asked whether the human PMP22 gene is a target for regulation by microRNA 29a (miR-29a). Using bioinformatics, we determined that the human PMP22 gene contains the conserved seed sequence of the miR-29a binding site and this regulatory motif is included in the duplicated region in neuropathic patients. Using luciferase reporter assays in HEK293 cells, we demonstrated that transient transfection of a miR-29a mimic is associated with reduction in PMP22 3'UTR reporter activity. Transfecting normal and humanized transgenic neuropathic mouse Schwann cells with a miR-29a expression plasmid effectively lowered both the endogenous mouse and the transgenic human PMP22 transcripts compared with control vector. In dermal fibroblasts derived from neuropathic patients, ectopic expression of miR-29a led to ~50% reduction in PMP22 mRNA, which corresponded to ~20% decrease in PMP22 protein levels. Significantly, miR-29a-mediated reduction in PMP22 mitigated the reduced mitotic capacity of the neuropathic cells. Together, these results support further testing of miR-29a and/or PMP22-targeting siRNAs as therapeutic agents for correcting the aberrant expression of PMP22 in neuropathic patients.
Collapse
Affiliation(s)
- Jacquelyn Serfecz
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Hannah Bazick
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Mohammed Omar Al Salihi
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Peter Turner
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Christopher Fields
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Rolf Renne
- Department of Molecular Genetics & Microbiology, College of Medicine University of Florida, Gainesville, FL, 32610, USA
- UF Health Cancer Center, College of Medicine University of Florida, Gainesville, FL, 32610, USA
| | - Lucia Notterpek
- Department of Neuroscience, College of Medicine University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
33
|
Prada V, Massucco S, Venturi C, Geroldi A, Bellone E, Mandich P, Minuto M, Varaldo E, Mancardi G, Grandis M, Schenone A. Diagnostic Value of Sural Nerve Biopsy: Retrospective Analysis of Clinical Cases From 1981 to 2017. Front Neurol 2019; 10:1218. [PMID: 31824401 PMCID: PMC6884026 DOI: 10.3389/fneur.2019.01218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/01/2019] [Indexed: 12/02/2022] Open
Abstract
Nerve biopsy represents the conclusive step in the diagnostic work-up of peripheral neuropathies, and its diagnostic yield is still debated. The aim of this study is to consider the impact of nerve biopsy on reaching a useful diagnosis in different peripheral neuropathies and its changing over time. We retrospectively analyzed 1,179 sural nerve biopsies performed in the period 1981–2017 at Neurological Clinic of Policlinico San Martino (Genoa). We relied on medical records and collected both clinical and pathological data in a database. Biopsy provided univocal diagnoses in 53% of cases (with an increase over time), multiple diagnostic options in 14%, while diagnosis was undetermined in 33% (undetermined reports decreased during the years). In 57% of patients, the pre-biopsy suspicion was confirmed, while in 43% sural biopsy modified the clinical diagnosis. The highest yield was in axonal neuropathies (29% undetermined reports vs. 40% in demyelinating and 48% in mixed neuropathies). In 68% of patients with vasculitic neuropathy, this etiology was already suspected, whereas in 32% nerve biopsy modified the clinical diagnosis. During the years, the number of annually performed biopsies decreased significantly (p = 0.007), with an increase in the mean age of patients (p < 0.0001). The percentage of hereditary neuropathies had a significant decrease (p = 0.016), while the rate of vasculitic and chronic inflammatory neuropathies increased (p < 0.0001). This is the largest Italian study addressing the yield of sural nerve biopsy. During the years, we observed a progressive refinement of the indication of this procedure, which confirms its utility for interstitial neuropathies, particularly if non-systemic vasculitic neuropathy is suspected.
Collapse
Affiliation(s)
- Valeria Prada
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy
| | - Sara Massucco
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy
| | - Consuelo Venturi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy
| | - Alessandro Geroldi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Department of Neurology, Policlinico San Martino IRCCS, Genoa, Italy
| | - Emilia Bellone
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Department of Neurology, Policlinico San Martino IRCCS, Genoa, Italy
| | - Paola Mandich
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Department of Neurology, Policlinico San Martino IRCCS, Genoa, Italy
| | - Michele Minuto
- Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy.,Department of Surgery, Policlinico San Martino IRCSS, Genoa, Italy
| | - Emanuela Varaldo
- Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy.,Department of Surgery, Policlinico San Martino IRCSS, Genoa, Italy
| | - Giovanni Mancardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Fondazione Maugeri ICS, Genova, Italy
| | - Marina Grandis
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Department of Neurology, Policlinico San Martino IRCCS, Genoa, Italy
| | - Angelo Schenone
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, Genoa, Italy.,Department of Neurology, Policlinico San Martino IRCCS, Genoa, Italy
| |
Collapse
|
34
|
Sahenk Z, Ozes B. Gene therapy to promote regeneration in Charcot-Marie-Tooth disease. Brain Res 2019; 1727:146533. [PMID: 31669284 DOI: 10.1016/j.brainres.2019.146533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
The molecular pathogenesis underlying Charcot-Marie-Tooth (CMT) neuropathy subtypes is becoming increasingly variable and identification of common approaches for treatment, independently of the disease causing gene defect, is therefore much desirable. Gene therapy approach from the clinical translational view point is particularly challenging for the most common "demyelinating" CMT1 subtypes, caused by primary Schwann cell genetic defects. Studies have shown that impaired regenerative capacity of distal axons is major contributing factor to distal axonal loss in primary Schwann cell genetic defects and neurotrophin 3 (NT-3) improves impaired regeneration in CMT1 mouse models. This review surveys the evidence supporting the rationale for AAV1.NT-3 surrogate gene therapy to improve nerve regeneration in CMT1A. The translational process, from proof of principal studies to the design of the phase I/IIa trial evaluating scAAV1.tMCK.NTF3 gene therapy for treatment of CMT1A is summarized.
Collapse
Affiliation(s)
- Zarife Sahenk
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States; Department of Pediatrics and Neurology, Nationwide Children's Hospital and The Ohio State University, United States; Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, United States; Department of Neurology, The Ohio State University, United States.
| | - Burcak Ozes
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
35
|
Pantera H, Shy ME, Svaren J. Regulating PMP22 expression as a dosage sensitive neuropathy gene. Brain Res 2019; 1726:146491. [PMID: 31586623 DOI: 10.1016/j.brainres.2019.146491] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Structural variation in the human genome has emerged as a major cause of disease as genomic data have accumulated. One of the most common structural variants associated with human disease causes the heritable neuropathy known as Charcot-Marie-Tooth (CMT) disease type 1A. This 1.4 Mb duplication causes nearly half of the CMT cases that are genetically diagnosed. The PMP22 gene is highly induced in Schwann cells during development, although its precise role in myelin formation and homeostasis is still under active investigation. The PMP22 gene can be considered as a nucleoprotein complex with enzymatic activity to produce the PMP22 transcript, and the complex is allosterically regulated by transcription factors that respond to intracellular signals and epigenomic modifications. The control of PMP22 transcript levels has been one of the major therapeutic targets of therapy development, and this review summarizes those approaches as well as efforts to characterize the regulation of the PMP22 gene.
Collapse
Affiliation(s)
- Harrison Pantera
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, WI, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
36
|
Chen C, Dong H, Wei Q, Li L, Yu H, Li J, Liu G, Li H, Bai G, Ma H, Wu Z. Genetic spectrum and clinical profiles in a southeast Chinese cohort of Charcot‐Marie‐Tooth disease. Clin Genet 2019; 96:439-448. [PMID: 31372974 DOI: 10.1111/cge.13616] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Cong‐Xin Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
- Department of Neurology and Institute of NeurologyFirst Affiliated Hospital, Fujian Medical University Fuzhou China
| | - Hai‐Lin Dong
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Qiao Wei
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Li‐Xi Li
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan University Shanghai China
| | - Hao Yu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Jia‐Qi Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Gong‐Lu Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Hong‐Fu Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Ge Bai
- Institute of Neuroscience and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Huan Ma
- Institute of Neuroscience and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| | - Zhi‐Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang ProvinceZhejiang University School of Medicine Hangzhou China
| |
Collapse
|
37
|
Nam SH, Choi BO. Clinical and genetic aspects of Charcot-Marie-Tooth disease subtypes. PRECISION AND FUTURE MEDICINE 2019. [DOI: 10.23838/pfm.2018.00163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
38
|
Tadenev ALD, Burgess RW. Model validity for preclinical studies in precision medicine: precisely how precise do we need to be? Mamm Genome 2019; 30:111-122. [PMID: 30953144 PMCID: PMC6606658 DOI: 10.1007/s00335-019-09798-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/27/2019] [Indexed: 12/15/2022]
Abstract
The promise of personalized medicine is that each patient’s treatment can be optimally tailored to their disease. In turn, their disease, as well as their response to the treatment, is determined by their genetic makeup and the “environment,” which relates to their general health, medical history, personal habits, and surroundings. Developing such optimized treatment strategies is an admirable goal and success stories include examples such as switching chemotherapy agents based on a patient’s tumor genotype. However, it remains a challenge to apply precision medicine to diseases for which there is no known effective treatment. Such diseases require additional research, often using experimentally tractable models. Presumably, models that recapitulate as much of the human pathophysiology as possible will be the most predictive. Here we will discuss the considerations behind such “precision models.” What sort of precision is required and under what circumstances? How can the predictive validity of such models be improved? Ultimately, there is no perfect model, but our continually improving ability to genetically engineer a variety of systems allows the generation of more and more precise models. Furthermore, our steadily increasing awareness of risk alleles, genetic background effects, multifactorial disease processes, and gene by environment interactions also allows increasingly sophisticated models that better reproduce patients’ conditions. In those cases where the research has progressed sufficiently far, results from these models appear to often be translating to effective treatments for patients.
Collapse
Affiliation(s)
- Abigail L D Tadenev
- The Center for Precision Genetics, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Robert W Burgess
- The Center for Precision Genetics, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
39
|
Kamil K, Yazid MD, Idrus RBH, Das S, Kumar J. Peripheral Demyelinating Diseases: From Biology to Translational Medicine. Front Neurol 2019; 10:87. [PMID: 30941082 PMCID: PMC6433847 DOI: 10.3389/fneur.2019.00087] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/22/2019] [Indexed: 12/30/2022] Open
Abstract
Demyelinating diseases represent a spectrum of disorders that impose significant burden on global economy and society. Generally, the prognosis of these diseases is poor and there is no available cure. In recent decades, research has shed some light on the biology and physiology of Schwann cells and its neuroprotective effects in the peripheral nervous system (PNS). Insults to the PNS by various infectious agents, genetic predisposition and immune-related mechanisms jeopardize Schwann cell functions and cause demyelination. To date, there are no effective and reliable biomarkers for PNS-related diseases. Here, we aim to review the following: pathogenesis of various types of peripheral demyelinating diseases such as Guillain-Barre syndrome, Chronic Inflammatory Demyelinating Polyradiculoneuropathy, Anti-Myelin Associated Glycoprotein Neuropathy, POEMS syndrome, and Charcot-Marie-Tooth disease; emerging novel biomarkers for peripheral demyelinating diseases, and Schwann cell associated markers for demyelination.
Collapse
Affiliation(s)
- Khidhir Kamil
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
40
|
Pantera H, Moran JJ, Hung HA, Pak E, Dutra A, Svaren J. Regulation of the neuropathy-associated Pmp22 gene by a distal super-enhancer. Hum Mol Genet 2019; 27:2830-2839. [PMID: 29771329 DOI: 10.1093/hmg/ddy191] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Peripheral nerve myelination is adversely affected in the most common form of the hereditary peripheral neuropathy called Charcot-Marie-Tooth Disease. This form, classified as CMT1A, is caused by a 1.4 Mb duplication on chromosome 17, which includes the abundantly expressed Schwann cell myelin gene, Peripheral Myelin Protein 22 (PMP22). This is one of the most common copy number variants causing neurological disease. Overexpression of Pmp22 in rodent models recapitulates several aspects of neuropathy, and reduction of Pmp22 in such models results in amelioration of the neuropathy phenotype. Recently we identified a potential super-enhancer approximately 90-130 kb upstream of the Pmp22 transcription start sites. This super-enhancer encompasses a cluster of individual enhancers that have the acetylated histone H3K27 active enhancer mark, and coincides with smaller duplications identified in patients with milder CMT1A-like symptoms, where the PMP22 coding region itself was not part of the duplication. In this study, we have utilized genome editing to create a deletion of this super-enhancer to determine its role in Pmp22 regulation. Our data show a significant decrease in Pmp22 transcript expression using allele-specific internal controls. Moreover, the P2 promoter of the Pmp22 gene, which is used in other cell types, is affected, but we find that the Schwann cell-specific P1 promoter is disproportionately more sensitive to loss of the super-enhancer. These data show for the first time the requirement of these upstream enhancers for full Pmp22 expression.
Collapse
Affiliation(s)
- Harrison Pantera
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA.,Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John J Moran
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Holly A Hung
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Evgenia Pak
- Cytogenetics and Microscopy Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amalia Dutra
- Cytogenetics and Microscopy Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
41
|
Prukop T, Stenzel J, Wernick S, Kungl T, Mroczek M, Adam J, Ewers D, Nabirotchkin S, Nave KA, Hajj R, Cohen D, Sereda MW. Early short-term PXT3003 combinational therapy delays disease onset in a transgenic rat model of Charcot-Marie-Tooth disease 1A (CMT1A). PLoS One 2019; 14:e0209752. [PMID: 30650121 PMCID: PMC6334894 DOI: 10.1371/journal.pone.0209752] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022] Open
Abstract
The most common type of Charcot-Marie-Tooth disease is caused by a duplication of PMP22 leading to dysmyelination, axonal loss and progressive muscle weakness (CMT1A). Currently, no approved therapy is available for CMT1A patients. A novel polytherapeutic proof-of-principle approach using PXT3003, a low-dose combination of baclofen, naltrexone and sorbitol, slowed disease progression after long-term dosing in adult Pmp22 transgenic rats, a known animal model of CMT1A. Here, we report an early postnatal, short-term treatment with PXT3003 in CMT1A rats that delays disease onset into adulthood. CMT1A rats were treated from postnatal day 6 to 18 with PXT3003. Behavioural, electrophysiological, histological and molecular analyses were performed until 12 weeks of age. Daily oral treatment for approximately 2 weeks ameliorated motor deficits of CMT1A rats reaching wildtype levels. Histologically, PXT3003 corrected the disturbed axon calibre distribution with a shift towards large motor axons. Despite dramatic clinical amelioration, only distal motor latencies were improved and correlated with phenotype performance. On the molecular level, PXT3003 reduced Pmp22 mRNA overexpression and improved the misbalanced downstream PI3K-AKT / MEK-ERK signalling pathway. The improved differentiation status of Schwann cells may have enabled better long-term axonal support function. We conclude that short-term treatment with PXT3003 during early development may partially prevent the clinical and molecular manifestations of CMT1A. Since PXT3003 has a strong safety profile and is currently undergoing a phase III trial in CMT1A patients, our results suggest that PXT3003 therapy may be a bona fide translatable therapy option for children and young adolescent patients suffering from CMT1A.
Collapse
Affiliation(s)
- Thomas Prukop
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Pharmacology, Göttingen, Germany
| | - Jan Stenzel
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Stephanie Wernick
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Theresa Kungl
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Magdalena Mroczek
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Julia Adam
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - David Ewers
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | | | - Klaus-Armin Nave
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | | | | | - Michael W. Sereda
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
- University Medical Center Göttingen, Department of Clinical Neurophysiology, Göttingen, Germany
- * E-mail:
| |
Collapse
|
42
|
Murakami T, Sunada Y. Schwann Cell and the Pathogenesis of Charcot–Marie–Tooth Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:301-321. [DOI: 10.1007/978-981-32-9636-7_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
43
|
Fledrich R, Abdelaal T, Rasch L, Bansal V, Schütza V, Brügger B, Lüchtenborg C, Prukop T, Stenzel J, Rahman RU, Hermes D, Ewers D, Möbius W, Ruhwedel T, Katona I, Weis J, Klein D, Martini R, Brück W, Müller WC, Bonn S, Bechmann I, Nave KA, Stassart RM, Sereda MW. Targeting myelin lipid metabolism as a potential therapeutic strategy in a model of CMT1A neuropathy. Nat Commun 2018; 9:3025. [PMID: 30072689 PMCID: PMC6072747 DOI: 10.1038/s41467-018-05420-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 06/28/2018] [Indexed: 01/17/2023] Open
Abstract
In patients with Charcot-Marie-Tooth disease 1A (CMT1A), peripheral nerves display aberrant myelination during postnatal development, followed by slowly progressive demyelination and axonal loss during adult life. Here, we show that myelinating Schwann cells in a rat model of CMT1A exhibit a developmental defect that includes reduced transcription of genes required for myelin lipid biosynthesis. Consequently, lipid incorporation into myelin is reduced, leading to an overall distorted stoichiometry of myelin proteins and lipids with ultrastructural changes of the myelin sheath. Substitution of phosphatidylcholine and phosphatidylethanolamine in the diet is sufficient to overcome the myelination deficit of affected Schwann cells in vivo. This treatment rescues the number of myelinated axons in the peripheral nerves of the CMT rats and leads to a marked amelioration of neuropathic symptoms. We propose that lipid supplementation is an easily translatable potential therapeutic approach in CMT1A and possibly other dysmyelinating neuropathies.
Collapse
Affiliation(s)
- R Fledrich
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Institute of Anatomy, University of Leipzig, Leipzig, 04103, Germany.
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany.
| | - T Abdelaal
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Division, National Research Centre, Giza, 12622, Egypt
| | - L Rasch
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - V Bansal
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - V Schütza
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany
| | - B Brügger
- Heidelberg University Biochemistry Center (BZH), Heidelberg, 69120, Germany
| | - C Lüchtenborg
- Heidelberg University Biochemistry Center (BZH), Heidelberg, 69120, Germany
| | - T Prukop
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - J Stenzel
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - R U Rahman
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - D Hermes
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - D Ewers
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - W Möbius
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, 37075, Germany
| | - T Ruhwedel
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany
| | - I Katona
- Institute of Neuropathology, University Hospital Aachen, Aachen, 52074, Germany
| | - J Weis
- Institute of Neuropathology, University Hospital Aachen, Aachen, 52074, Germany
| | - D Klein
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Wuerzburg, Wuerzburg, 97080, Germany
| | - R Martini
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Wuerzburg, Wuerzburg, 97080, Germany
| | - W Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - W C Müller
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany
| | - S Bonn
- Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
- German Center for Neurodegenerative Diseases, Tübingen, 72076, Germany
| | - I Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, 04103, Germany
| | - K A Nave
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
| | - R M Stassart
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Department of Neuropathology, University Hospital Leipzig, Leipzig, 04103, Germany.
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| | - M W Sereda
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, 37075, Germany.
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| |
Collapse
|
44
|
Law WD, Fogarty EA, Vester A, Antonellis A. A genome-wide assessment of conserved SNP alleles reveals a panel of regulatory SNPs relevant to the peripheral nerve. BMC Genomics 2018; 19:311. [PMID: 29716548 PMCID: PMC5930951 DOI: 10.1186/s12864-018-4692-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 04/17/2018] [Indexed: 12/29/2022] Open
Abstract
Background Identifying functional non-coding variation is critical for defining the genetic contributions to human disease. While single-nucleotide polymorphisms (SNPs) within cis-acting transcriptional regulatory elements have been implicated in disease pathogenesis, not all cell types have been assessed and functional validations have been limited. In particular, the cells of the peripheral nervous system have been excluded from genome-wide efforts to link non-coding SNPs to altered gene function. Addressing this gap is essential for defining the genetic architecture of diseases that affect the peripheral nerve. We developed a computational pipeline to identify SNPs that affect regulatory function (rSNPs) and evaluated our predictions on a set of 144 regions in Schwann cells, motor neurons, and muscle cells. Results We identified 28 regions that display regulatory activity in at least one cell type and 13 SNPs that affect regulatory function. We then tailored our pipeline to one peripheral nerve cell type by incorporating SOX10 ChIP-Seq data; SOX10 is essential for Schwann cells. We prioritized 22 putative SOX10 response elements harboring a SNP and rapidly validated two rSNPs. We then selected one of these elements for further characterization to assess the biological relevance of our approach. Deletion of the element from the genome of cultured Schwann cells—followed by differential gene expression studies—revealed Tubb2b as a candidate target gene. Studying the enhancer in developing mouse embryos revealed activity in SOX10-positive cells including the dorsal root ganglia and melanoblasts. Conclusions Our efforts provide insight into the utility of employing strict conservation for rSNP discovery. This strategy, combined with functional analyses, can yield candidate target genes. In support of this, our efforts suggest that investigating the role of Tubb2b in SOX10-positive cells may reveal novel biology within these cell populations. Electronic supplementary material The online version of this article (10.1186/s12864-018-4692-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Law
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elizabeth A Fogarty
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aimée Vester
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anthony Antonellis
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA. .,Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Neurology, University of Michigan Medical School, 3710A Medical Sciences II, 1241 E. Catherine St. SPC 5618, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
45
|
Kiepura AJ, Kochański A. Charcot-Marie-Tooth type 1A drug therapies: role of adenylyl cyclase activity and G-protein coupled receptors in disease pathomechanism. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
46
|
Duchesne M, Mathis S, Richard L, Magdelaine C, Corcia P, Nouioua S, Tazir M, Magy L, Vallat JM. Nerve Biopsy Is Still Useful in Some Inherited Neuropathies. J Neuropathol Exp Neurol 2017; 77:88-99. [DOI: 10.1093/jnen/nlx111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
47
|
Zhao HT, Damle S, Ikeda-Lee K, Kuntz S, Li J, Mohan A, Kim A, Hung G, Scheideler MA, Scherer SS, Svaren J, Swayze EE, Kordasiewicz HB. PMP22 antisense oligonucleotides reverse Charcot-Marie-Tooth disease type 1A features in rodent models. J Clin Invest 2017; 128:359-368. [PMID: 29202483 DOI: 10.1172/jci96499] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/03/2017] [Indexed: 11/17/2022] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is caused by duplication of peripheral myelin protein 22 (PMP22) and is the most common hereditary peripheral neuropathy. CMT1A is characterized by demyelination and axonal loss, which underlie slowed motor nerve conduction velocity (MNCV) and reduced compound muscle action potentials (CMAP) in patients. There is currently no known treatment for this disease. Here, we show that antisense oligonucleotides (ASOs) effectively suppress PMP22 mRNA in affected nerves in 2 murine CMT1A models. Notably, initiation of ASO treatment after disease onset restored myelination, MNCV, and CMAP almost to levels seen in WT animals. In addition to disease-associated gene expression networks that were restored with ASO treatment, we also identified potential disease biomarkers through transcriptomic profiling. Furthermore, we demonstrated that reduction of PMP22 mRNA in skin biopsies from ASO-treated rats is a suitable biomarker for evaluating target engagement in response to ASO therapy. These results support the use of ASOs as a potential treatment for CMT1A and elucidate potential disease and target engagement biomarkers for use in future clinical trials.
Collapse
Affiliation(s)
| | - Sagar Damle
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | | | - Steven Kuntz
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | - Jian Li
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Apoorva Mohan
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | - Aneeza Kim
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | - Gene Hung
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | | | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eric E Swayze
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | | |
Collapse
|
48
|
Fledrich R, Mannil M, Leha A, Ehbrecht C, Solari A, Pelayo-Negro AL, Berciano J, Schlotter-Weigel B, Schnizer TJ, Prukop T, Garcia-Angarita N, Czesnik D, Haberlová J, Mazanec R, Paulus W, Beissbarth T, Walter MC, CMT-TRIAAL, Hogrel JY, Dubourg O, Schenone A, Baets J, De Jonghe P, Shy ME, Horvath R, Pareyson D, Seeman P, Young P, Sereda MW. Biomarkers predict outcome in Charcot-Marie-Tooth disease 1A. J Neurol Neurosurg Psychiatry 2017; 88:941-952. [PMID: 28860329 PMCID: PMC8265963 DOI: 10.1136/jnnp-2017-315721] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/05/2017] [Accepted: 05/02/2017] [Indexed: 11/03/2022]
Abstract
BACKGROUND Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited neuropathy, a debilitating disease without known cure. Among patients with CMT1A, disease manifestation, progression and severity are strikingly variable, which poses major challenges for the development of new therapies. Hence, there is a strong need for sensitive outcome measures such as disease and progression biomarkers, which would add powerful tools to monitor therapeutic effects in CMT1A. METHODS We established a pan-European and American consortium comprising nine clinical centres including 311 patients with CMT1A in total. From all patients, the CMT neuropathy score and secondary outcome measures were obtained and a skin biopsy collected. In order to assess and validate disease severity and progression biomarkers, we performed qPCR on a set of 16 animal model-derived potential biomarkers in skin biopsy mRNA extracts. RESULTS In 266 patients with CMT1A, a cluster of eight cutaneous transcripts differentiates disease severity with a sensitivity and specificity of 90% and 76.1%, respectively. In an additional cohort of 45 patients with CMT1A, from whom a second skin biopsy was taken after 2-3 years, the cutaneous mRNA expression of GSTT2, CTSA, PPARG, CDA, ENPP1 and NRG1-Iis changing over time and correlates with disease progression. CONCLUSIONS In summary, we provide evidence that cutaneous transcripts in patients with CMT1A serve as disease severity and progression biomarkers and, if implemented into clinical trials, they could markedly accelerate the development of a therapy for CMT1A.
Collapse
Affiliation(s)
- Robert Fledrich
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Research Group “Molecular and Translational Neurology”, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Manoj Mannil
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Research Group “Molecular and Translational Neurology”, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Andreas Leha
- Department of Medical Statistics, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Caroline Ehbrecht
- Research Group “Molecular and Translational Neurology”, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Alessandra Solari
- Unit of Neuroepidemiology, IRCCS Foundation, C. Besta Neurological Institute, Milan, Italy
| | - Ana L. Pelayo-Negro
- Service of Neurology, University Hospital “Marqués de Valdecilla (IDIVAL)”, University of Cantabria, and “Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)”, Santander, Spain
| | - José Berciano
- Service of Neurology, University Hospital “Marqués de Valdecilla (IDIVAL)”, University of Cantabria, and “Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)”, Santander, Spain
| | - Beate Schlotter-Weigel
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University of Munich, Germany
| | - Tuuli J. Schnizer
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Thomas Prukop
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Research Group “Molecular and Translational Neurology”, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Institute of Clinical Pharmacology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Natalia Garcia-Angarita
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University of Munich, Germany
| | - Dirk Czesnik
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Haberlová
- Department of Child Neurology, Charles University in Prague, 2nd Medical School, and University Hospital Motol Prague, Czech Republic
| | - Radim Mazanec
- Department of Child Neurology, Charles University in Prague, 2nd Medical School, and University Hospital Motol Prague, Czech Republic
| | - Walter Paulus
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Tim Beissbarth
- Department of Medical Statistics, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Maggie C. Walter
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University of Munich, Germany
| | - CMT-TRIAAL
- CMT-TRIAAL (all participants in the appendix of this manuscript)
| | | | - Odile Dubourg
- Institute of Myology, GH Pitié-Salpêtrière, Paris, France
| | - Angelo Schenone
- Department of Neurology, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | - Jonathan Baets
- Neurogenetics Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerpen, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerpen, Belgium
| | - Peter De Jonghe
- Neurogenetics Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerpen, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerpen, Belgium
| | - Michael E. Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Rita Horvath
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, UK
| | - Davide Pareyson
- Unit of Neurological Rare Diseases of Adulthood, Department of Clinical Neurosciences, IRCCS Foundation, C. Besta Neurological Institute, Milan, Italy
| | - Pavel Seeman
- Department of Child Neurology, Charles University in Prague, 2nd Medical School, and University Hospital Motol Prague, Czech Republic
| | - Peter Young
- Department of Sleep Medicine and Neuromuscular Disorders, University Hospital Münster, Germany
| | - Michael W. Sereda
- Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Research Group “Molecular and Translational Neurology”, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
49
|
Prior R, Van Helleputte L, Benoy V, Van Den Bosch L. Defective axonal transport: A common pathological mechanism in inherited and acquired peripheral neuropathies. Neurobiol Dis 2017; 105:300-320. [DOI: 10.1016/j.nbd.2017.02.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/29/2017] [Accepted: 02/20/2017] [Indexed: 12/29/2022] Open
|
50
|
Soldevilla B, Cuevas-Martín C, Ibáñez C, Santacatterina F, Alberti MA, Simó C, Casasnovas C, Márquez-Infante C, Sevilla T, Pascual SI, Sánchez-Aragó M, Espinos C, Palau F, Cuezva JM. Plasma metabolome and skin proteins in Charcot-Marie-Tooth 1A patients. PLoS One 2017; 12:e0178376. [PMID: 28575008 PMCID: PMC5456076 DOI: 10.1371/journal.pone.0178376] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/11/2017] [Indexed: 12/01/2022] Open
Abstract
Objective Charcot-Marie-Tooth 1A (CMT1A) disease is the most common inherited neuropathy that lacks of therapy and of molecular markers to assess disease severity. Herein, we have pursued the identification of potential biomarkers in plasma samples and skin biopsies that could define the phenotype of CMT1A patients at mild (Mi), moderate (Mo) and severe (Se) stages of disease as assessed by the CMT neuropathy score to contribute to the understanding of CMT pathophysiology and eventually inform of the severity of the disease. Methods We have used: (i) a high-throughput untargeted metabolomic approach of plasma samples in a cohort of 42 CMT1A patients and 15 healthy controls (CRL) using ultrahigh liquid chromatography coupled to mass spectrometry and (ii) reverse phase protein microarrays to quantitate the expression of some proteins of energy metabolism and of the antioxidant response in skin biopsies of a cohort of 70 CMT1A patients and 13 healthy controls. Results The metabolomic approach identified 194 metabolites with significant differences among the four groups (Mi, Mo, Se, CRL) of samples. A multivariate Linear Discriminant Analysis model using 12 metabolites afforded the correct classification of the samples. These metabolites indicate an increase in protein catabolism and the mobilization of membrane lipids involved in signaling inflammation with severity of CMT1A. A concurrent depletion of leucine, which is required for the biogenesis of the muscle, is also observed in the patients. Protein expression in skin biopsies indicates early loss of mitochondrial and antioxidant proteins in patients’ biopsies. Conclusion The findings indicate that CMT1A disease is associated with a metabolic state resembling inflammation and sarcopenia suggesting that it might represent a potential target to prevent the nerve and muscle wasting phenotype in these patients. The observed changes in metabolites could be useful as potential biomarkers of CMT1A disease after appropriate validation in future longitudinal studies.
Collapse
Affiliation(s)
- Beatriz Soldevilla
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Cuevas-Martín
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Clara Ibáñez
- Instituto de Investigación en Ciencias de la Alimentación, Consejo Superior de Investigaciones Científicas (CIAL-CSIC), Madrid, Spain
- Nutritional Genomics and Food GENYAL Platform, IMDEA Food Institute, Madrid, Spain
| | - Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - María A. Alberti
- Unidad Neuromuscular, IIS Hospital Universitario de Bellvitge, IDIBELL, l’Hospitalet de Llobegrat, Spain
| | - Carolina Simó
- Instituto de Investigación en Ciencias de la Alimentación, Consejo Superior de Investigaciones Científicas (CIAL-CSIC), Madrid, Spain
| | - Carlos Casasnovas
- Unidad Neuromuscular, IIS Hospital Universitario de Bellvitge, IDIBELL, l’Hospitalet de Llobegrat, Spain
| | - Celedonio Márquez-Infante
- Servicio de Neurología y Neurofisiología, IIS Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Teresa Sevilla
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- IIS Hospital Universitari i Politecnic La Fe, Departamento de Medicina, Universidad de Valencia, Valencia, Spain
| | | | - María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Espinos
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Francesc Palau
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Division of Pediatrics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|