1
|
Sumner CJ, Miller TM. The expanding application of antisense oligonucleotides to neurodegenerative diseases. J Clin Invest 2024; 134:e186116. [PMID: 39352381 PMCID: PMC11444189 DOI: 10.1172/jci186116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024] Open
Affiliation(s)
- Charlotte J. Sumner
- Departments of Neurology, Neuroscience, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore Maryland, USA
| | - Timothy M. Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Dong H, Qin B, Zhang H, Lei L, Wu S. Current Treatment Methods for Charcot-Marie-Tooth Diseases. Biomolecules 2024; 14:1138. [PMID: 39334903 PMCID: PMC11430469 DOI: 10.3390/biom14091138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Charcot-Marie-Tooth (CMT) disease, the most common inherited neuromuscular disorder, exhibits a wide phenotypic range, genetic heterogeneity, and a variable disease course. The diverse molecular genetic mechanisms of CMT were discovered over the past three decades with the development of molecular biology and gene sequencing technologies. These methods have brought new options for CMT reclassification and led to an exciting era of treatment target discovery for this incurable disease. Currently, there are no approved disease management methods that can fully cure patients with CMT, and rehabilitation, orthotics, and surgery are the only available treatments to ameliorate symptoms. Considerable research attention has been given to disease-modifying therapies, including gene silencing, gene addition, and gene editing, but most treatments that reach clinical trials are drug treatments, while currently, only gene therapies for CMT2S have reached the clinical trial stage. In this review, we highlight the pathogenic mechanisms and therapeutic investigations of different subtypes of CMT, and promising therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Hongxian Dong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (H.D.); (B.Q.); (H.Z.)
| | - Boquan Qin
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (H.D.); (B.Q.); (H.Z.)
| | - Hui Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (H.D.); (B.Q.); (H.Z.)
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shizhou Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (H.D.); (B.Q.); (H.Z.)
| |
Collapse
|
3
|
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
|
4
|
McCulloch MK, Mehryab F, Rashnonejad A. Navigating the Landscape of CMT1B: Understanding Genetic Pathways, Disease Models, and Potential Therapeutic Approaches. Int J Mol Sci 2024; 25:9227. [PMID: 39273178 PMCID: PMC11395143 DOI: 10.3390/ijms25179227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Charcot-Marie-Tooth type 1B (CMT1B) is a peripheral neuropathy caused by mutations in the gene encoding myelin protein zero (MPZ), a key component of the myelin sheath in Schwann cells. Mutations in the MPZ gene can lead to protein misfolding, unfolded protein response (UPR), endoplasmic reticulum (ER) stress, or protein mistrafficking. Despite significant progress in understanding the disease mechanisms, there is currently no effective treatment for CMT1B, with therapeutic strategies primarily focused on supportive care. Gene therapy represents a promising therapeutic approach for treating CMT1B. To develop a treatment and better design preclinical studies, an in-depth understanding of the pathophysiological mechanisms and animal models is essential. In this review, we present a comprehensive overview of the disease mechanisms, preclinical models, and recent advancements in therapeutic research for CMT1B, while also addressing the existing challenges in the field. This review aims to deepen the understanding of CMT1B and to encourage further research towards the development of effective treatments for CMT1B patients.
Collapse
Affiliation(s)
- Mary Kate McCulloch
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Fatemeh Mehryab
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
| | - Afrooz Rashnonejad
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
5
|
De Grado A, Pisciotta C, Saveri P, Pareyson D. Will new investigational drugs change the way we treat Charcot-Marie-Tooth disease? Expert Opin Investig Drugs 2024; 33:653-656. [PMID: 38712465 DOI: 10.1080/13543784.2024.2352635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Affiliation(s)
- Amedeo De Grado
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pisciotta
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Saveri
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
6
|
Scherer SS, Svaren J. Peripheral Nervous System (PNS) Myelin Diseases. Cold Spring Harb Perspect Biol 2024; 16:a041376. [PMID: 38253417 PMCID: PMC11065170 DOI: 10.1101/cshperspect.a041376] [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: 01/24/2024]
Abstract
This is a review of inherited and acquired causes of human demyelinating neuropathies and a subset of disorders that affect axon-Schwann cell interactions. Nearly all inherited demyelinating neuropathies are caused by mutations in genes that are expressed by myelinating Schwann cells, affecting diverse functions in a cell-autonomous manner. The most common acquired demyelinating neuropathies are Guillain-Barré syndrome and chronic, inflammatory demyelinating polyneuropathy, both of which are immune-mediated. An additional group of inherited and acquired disorders affect axon-Schwann cell interactions in the nodal region. Overall, these disorders affect the formation of myelin and its maintenance, with superimposed axonal loss that is clinically important.
Collapse
Affiliation(s)
- Steven S Scherer
- Department of Neurology, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - John Svaren
- Department of Comparative Biosciences, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
7
|
Sivera Mascaró R, García Sobrino T, Horga Hernández A, Pelayo Negro AL, Alonso Jiménez A, Antelo Pose A, Calabria Gallego MD, Casasnovas C, Cemillán Fernández CA, Esteban Pérez J, Fenollar Cortés M, Frasquet Carrera M, Gallano Petit MP, Giménez Muñoz A, Gutiérrez Gutiérrez G, Gutiérrez Martínez A, Juntas Morales R, Ciano-Petersen NL, Martínez Ulloa PL, Mederer Hengstl S, Millet Sancho E, Navacerrada Barrero FJ, Navarrete Faubel FE, Pardo Fernández J, Pascual Pascual SI, Pérez Lucas J, Pino Mínguez J, Rabasa Pérez M, Sánchez González M, Sotoca J, Rodríguez Santiago B, Rojas García R, Turon-Sans J, Vicent Carsí V, Sevilla Mantecón T. Clinical practice guidelines for the diagnosis and management of Charcot-Marie-Tooth disease. Neurologia 2024:S2173-5808(24)00047-6. [PMID: 38431252 DOI: 10.1016/j.nrleng.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Charcot-Marie-Tooth (CMT) disease is classified considering the neurophysiological and histological findings, the inheritance pattern and the underlying genetic defect. In recent years, with the advent of next generation sequencing, genetic complexity has increased exponentially, expanding the knowledge about disease pathways, and having an impact in clinical management. The aim of this guide is to offer recommendations for the diagnosis, prognosis, monitoring and treatment of this disease in Spain. MATERIAL AND METHODS This consensus guideline has been developed by a multidisciplinary panel encompassing a broad group of professionals including neurologists, neuropediatricians, geneticists, rehabilitators, and orthopaedic surgeons. RECOMMENDATIONS The diagnosis is based in the clinical characterization, usually presenting with a common phenotype. It should be followed by an appropriate neurophysiological study that allows for a correct classification, specific recommendations are established for the parameters that should be included. Genetic diagnosis must be approached in sequentially, once the PMP22 duplication has been ruled out if appropriate, a next generation sequencing should be considered taking into account the limitations of the available techniques. To date, there is no pharmacological treatment that modifies the course of the disease, but symptomatic management is important, as are the rehabilitation and orthopaedic considerations. The latter should be initiated early to identify and improve the patient's functional impairments, including individualised exercise guidelines, orthotic adaptation, and assessment of conservative surgeries such as tendon transpositions. The follow-up of patients with CMT is exclusively clinical, ancillary testing are not necessary in routine clinical practice.
Collapse
Affiliation(s)
- R Sivera Mascaró
- Servicio de Neurología, Hospital Universitari i Politécnic La Fe, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - T García Sobrino
- Servicio de Neurología, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain.
| | - A Horga Hernández
- Servicio de Neurología, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - A L Pelayo Negro
- Servicio de Neurología, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Center for Biomedical Research in the Neurodegenerative Diseases (CIBERNED) Network, Madrid, Spain
| | - A Alonso Jiménez
- Neuromuscular Reference Center, Neurology Department, University Hospital of Antwerp, Amberes, Belgium
| | - A Antelo Pose
- Servicio de Rehabilitación, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
| | | | - C Casasnovas
- Unitat de Neuromuscular, Servicio de Neurología, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | | | - J Esteban Pérez
- Servicio de Neurología, Unidad de ELA y Enfermedades Neuromusculares, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M Fenollar Cortés
- Genética Clínica, Servicio de Análisis Clínicos, Instituto de Medicina del Laboratorio, IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| | - M Frasquet Carrera
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Neurología, Hospital Universitari Dr. Peset, Valencia, Spain
| | - M P Gallano Petit
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - A Giménez Muñoz
- Servicio de Neurología, Hospital Royo Villanova, Zaragoza, Spain
| | - G Gutiérrez Gutiérrez
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Neurología, Hospital Universitario Infanta Sofía, San Sebastián de los Reyes, Madrid, Spain; Facultad de Medicina, Universidad Europea de Madrid, Madrid, Spain
| | - A Gutiérrez Martínez
- Servicio de Neurología, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - R Juntas Morales
- Servicio de Neurología, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - N L Ciano-Petersen
- Servicio de Neurología, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - P L Martínez Ulloa
- Servicio de Neurología, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - S Mederer Hengstl
- Servicio de Neurología, Complejo Hospitalario de Pontevedra, Pontevedra, Spain
| | - E Millet Sancho
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Neurofisiología, Hospital Universitari i Politécnic La Fe, Instituto de Investigación Sanitaria la Fe, Valencia, Spain
| | - F J Navacerrada Barrero
- Servicio de Neurología, Hospital Universitario Infanta Sofía, San Sebastián de los Reyes, Madrid, Spain
| | - F E Navarrete Faubel
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Universitari i Politécnic La Fe, Valencia, Spain
| | - J Pardo Fernández
- Servicio de Neurología, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
| | | | - J Pérez Lucas
- Servicio de Neurología, Hospital del Tajo, Aranjuez, Madrid, Spain
| | - J Pino Mínguez
- Servicio de Cirugía Ortopédica y Traumatología, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, Spain
| | - M Rabasa Pérez
- Servicio de Neurología, Hospital Universitario de Fuenlabrada, Fuenlabrada, Madrid, Spain
| | - M Sánchez González
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Universitari i Politécnic La Fe, Valencia, Spain
| | - J Sotoca
- Servicio de Neurología, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - R Rojas García
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Neurología, Hospital de la Santa Creu i Sant Pau, Departamento de Medicina, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - J Turon-Sans
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Servicio de Neurofisiología, Hospital de la Santa Creu i Sant Pau, Departamento de Medicina, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - V Vicent Carsí
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Universitari i Politécnic La Fe, Valencia, Spain
| | - T Sevilla Mantecón
- Servicio de Neurología, Hospital Universitari i Politécnic La Fe, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Madrid, Spain; Universidad de Valencia, Valencia, Spain
| |
Collapse
|
8
|
Mandarakas MR, Eichinger KJ, Bray P, Cornett KMD, Shy ME, Reilly MM, Ramdharry GM, Scherer SS, Pareyson D, Estilow T, McKay MJ, Herrmann DN, Burns J. Multicenter Validation of the Charcot-Marie-Tooth Functional Outcome Measure. Neurology 2024; 102:e207963. [PMID: 38237108 PMCID: PMC11097760 DOI: 10.1212/wnl.0000000000207963] [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: 05/09/2023] [Accepted: 09/13/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Charcot-Marie-Tooth disease type 1A (CMT1A), caused by a duplication of PMP22, is the most common hereditary peripheral neuropathy. For participants with CMT1A, few clinical trials have been performed; however, multiple therapies have reached an advanced stage of preclinical development. In preparation for imminent clinical trials in participants with CMT1A, we have produced a Clinical Outcome Assessment (COA), known as the CMT-Functional Outcome Measure (CMT-FOM), in accordance with the FDA Roadmap to Patient-Focused Outcome Measurement to capture the key clinical end point of function. METHODS Participants were recruited through CMT clinics in the United States (n = 130), the United Kingdom (n = 52), and Italy (n = 32). To derive the most accurate signal with the fewest items to identify a therapeutic response, a series of validation studies were conducted including item and factor analysis, Rasch model analysis and testing of interrater reliability, discriminative ability, and convergent validity. RESULTS A total of 214 participants aged 18-75 years with CMT1A (58% female) were included in this study. Item, factor, and Rasch analysis supported the viability of the 12-item CMT-FOM as a unidimensional interval scale of function in adults with CMT1A. The CMT-FOM covers strength, upper and lower limb function, balance, and mobility. The 0-100 point scoring system showed good overall model fit, no evidence of misfitting items, and no person misfit, and it was well targeted for adults with CMT1A exhibiting high inter-rater reliability across a range of clinical settings and evaluators. The CMT-FOM was significantly correlated with the CMT Examination Score (r = 0.643; p < 0.001) and the Overall Neuropathy Limitation Scale (r = 0.516; p < 0.001). Significantly higher CMT-FOM total scores were observed in participants self-reporting daily trips and falls, unsteady ankles, hand tremor, and hand weakness (p < 0.05). DISCUSSION The CMT-FOM is a psychometrically robust multi-item, unidimensional, disease-specific COA covering strength, upper and lower limb function, balance, and mobility to capture how participants with CMT1A function to identify therapeutic efficacy.
Collapse
Affiliation(s)
- Melissa R Mandarakas
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Katy J Eichinger
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Paula Bray
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Kayla M D Cornett
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Michael E Shy
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Mary M Reilly
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Gita M Ramdharry
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Steven S Scherer
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Davide Pareyson
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Timothy Estilow
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Marnee J McKay
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - David N Herrmann
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| | - Joshua Burns
- From the The University of Sydney School of Health Sciences (M.R.M., P.B., K.M.D.C., M.J.M., J.B.), Faculty of Medicine and Health; Sydney Children's Hospitals Network (Randwick and Westmead) (M.R.M., P.B., K.M.C., J.B.), New South Wales, Australia; Department of Neurology (K.J.E., D.N.H.), University of Rochester, NY; Department of Neurology (M.E.S.), Carver College of Medicine, University of Iowa; Centre for Neuromuscular Diseases (M.M.R., G.M.R.), Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology (S.S.S.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Fondazione IRCCS Istituto Neurologico Carlo Besta (D.P.), Milan, Italy; and The Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania (T.E.), Philadelphia
| |
Collapse
|
9
|
Zeng S, Yang H, Wang B, Xie Y, Xu K, Liu L, Cao W, Liu X, Tang B, Liu M, Zhang R. The MORC2 p.S87L mutation reduces proliferation of pluripotent stem cells derived from a patient with the spinal muscular atrophy-like phenotype by inhibiting proliferation-related signaling pathways. Neural Regen Res 2024; 19:205-211. [PMID: 37488868 PMCID: PMC10479865 DOI: 10.4103/1673-5374.375347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 07/26/2023] Open
Abstract
Mutations in the microrchidia CW-type zinc finger protein 2 (MORC2) gene are the causative agent of Charcot-Marie-Tooth disease type 2Z (CMT2Z), and the hotspot mutation p.S87L is associated with a more severe spinal muscular atrophy-like clinical phenotype. The aims of this study were to determine the mechanism of the severe phenotype caused by the MORC2 p.S87L mutation and to explore potential treatment strategies. Epithelial cells were isolated from urine samples from a spinal muscular atrophy (SMA)-like patient (MORC2 p.S87L), a CMT2Z patient (MORC2 p.Q400R), and a healthy control and induced to generate pluripotent stem cells, which were then differentiated into motor neuron precursor cells. Next-generation RNA sequencing followed by KEGG pathway enrichment analysis revealed that differentially expressed genes involved in the PI3K/Akt and MAPK/ERK signaling pathways were enriched in the p.S87L SMA-like patient group and were significantly downregulated in induced pluripotent stem cells. Reduced proliferation was observed in the induced pluripotent stem cells and motor neuron precursor cells derived from the p.S87L SMA-like patient group compared with the CMT2Z patient group and the healthy control. G0/G1 phase cell cycle arrest was observed in induced pluripotent stem cells derived from the p.S87L SMA-like patient. MORC2 p.S87L-specific antisense oligonucleotides (p.S87L-ASO-targeting) showed significant efficacy in improving cell proliferation and activating the PI3K/Akt and MAPK/ERK pathways in induced pluripotent stem cells. However, p.S87L-ASO-targeting did not rescue proliferation of motor neuron precursor cells. These findings suggest that downregulation of the PI3K/Akt and MAPK/ERK signaling pathways leading to reduced cell proliferation and G0/G1 phase cell cycle arrest in induced pluripotent stem cells might be the underlying mechanism of the severe p.S87L SMA-like phenotype. p.S87L-ASO-targeting treatment can alleviate disordered cell proliferation in the early stage of pluripotent stem cell induction.
Collapse
Affiliation(s)
- Sen Zeng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Honglan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Binghao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ke Xu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lei Liu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wanqian Cao
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Beisha Tang
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan Province, China
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
10
|
Nam YH, Park S, Yum Y, Jeong S, Park HE, Kim HJ, Lim J, Choi BO, Jung SC. Preclinical Efficacy of Peripheral Nerve Regeneration by Schwann Cell-like Cells Differentiated from Human Tonsil-Derived Mesenchymal Stem Cells in C22 Mice. Biomedicines 2023; 11:3334. [PMID: 38137555 PMCID: PMC10741921 DOI: 10.3390/biomedicines11123334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is a hereditary disease with heterogeneous phenotypes and genetic causes. CMT type 1A (CMT1A) is a type of disease affecting the peripheral nerves and is caused by the duplication of the peripheral myelin protein 22 (PMP22) gene. Human tonsil-derived mesenchymal stem cells (TMSCs) are useful for stem cell therapy in various diseases and can be differentiated into Schwann cell-like cells (TMSC-SCs). We investigated the potential of TMSC-SCs called neuronal regeneration-promoting cells (NRPCs) for peripheral nerve and muscle regeneration in C22 mice, a model for CMT1A. We transplanted NRPCs manufactured in a good manufacturing practice facility into the bilateral thigh muscles of C22 mice and performed behavior and nerve conduction tests and histological and ultrastructural analyses. Significantly, the motor function was much improved, the ratio of myelinated axons was increased, and the G-ratio was reduced by the transplantation of NRPCs. The sciatic nerve and gastrocnemius muscle regeneration of C22 mice following the transplantation of NRPCs downregulated PMP22 overexpression, which was observed in a dose-dependent manner. These results suggest that NRPCs are feasible for clinical research for the treatment of CMT1A patients. Research applying NRPCs to other peripheral nerve diseases is also needed.
Collapse
Affiliation(s)
- Yu Hwa Nam
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (Y.H.N.); (S.P.); (Y.Y.); (S.J.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Saeyoung Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (Y.H.N.); (S.P.); (Y.Y.); (S.J.)
| | - Yoonji Yum
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (Y.H.N.); (S.P.); (Y.Y.); (S.J.)
| | - Soyeon Jeong
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (Y.H.N.); (S.P.); (Y.Y.); (S.J.)
| | - Hyo Eun Park
- Cellatoz Therapeutics Inc., Seongnam-si 13487, Gyeonggi-do, Republic of Korea; (H.E.P.); (H.J.K.); (J.L.)
| | - Ho Jin Kim
- Cellatoz Therapeutics Inc., Seongnam-si 13487, Gyeonggi-do, Republic of Korea; (H.E.P.); (H.J.K.); (J.L.)
| | - Jaeseung Lim
- Cellatoz Therapeutics Inc., Seongnam-si 13487, Gyeonggi-do, Republic of Korea; (H.E.P.); (H.J.K.); (J.L.)
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Sung-Chul Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (Y.H.N.); (S.P.); (Y.Y.); (S.J.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
11
|
Canalis E, Mocarska M, Schilling L, Jafar-Nejad P, Carrer M. Antisense oligonucleotides targeting a NOTCH3 mutation in male mice ameliorate the cortical osteopenia of lateral meningocele syndrome. Bone 2023; 177:116898. [PMID: 37704069 PMCID: PMC10591917 DOI: 10.1016/j.bone.2023.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
Lateral Meningocele Syndrome (LMS) is a monogenic disorder associated with NOTCH3 pathogenic variants that result in the stabilization of NOTCH3 and a gain-of-function. A mouse model (Notch3em1Ecan) harboring a 6691-TAATGA mutation in the Notch3 locus that results in a functional outcome analogous to LMS exhibits cancellous and cortical bone osteopenia. We tested Notch3 antisense oligonucleotides (ASOs) specific to the Notch36691-TAATGA mutation for their effects on Notch3 downregulation and on the osteopenia of Notch3em1Ecan mice. Twenty-four mouse Notch3 mutant ASOs were designed and tested for toxic effects in vivo, and 12 safe ASOs were tested for their impact on the downregulation of Notch36691-TAATGA and Notch3 mRNA in osteoblast cultures from Notch3em1Ecan mice. Three ASOs downregulated Notch3 mutant transcripts specifically and were tested in vivo for their effects on the bone microarchitecture of Notch3em1Ecan mice. All three ASOs were well tolerated. One of these ASOs had more consistent effects in vivo and was studied in detail. The Notch3 mutant ASO downregulated Notch3 mutant transcripts in osteoblasts and bone marrow stromal cells and had no effect on other Notch receptors. The subcutaneous administration of Notch3 mutant ASO at 50 mg/Kg decreased Notch36691-TAATGA mRNA in bone without apparent toxicity; microcomputed tomography demonstrated that the ASO ameliorated the cortical osteopenia of Notch3em1Ecan mice but not the cancellous bone osteopenia. In conclusion, a Notch3 ASO that downregulates Notch3 mutant expression specifically ameliorates the cortical osteopenia in Notch3em1Ecan mice. ASOs may become useful strategies in the management of monogenic disorders affecting the skeleton.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA; Department of Medicine, UConn Health, Farmington, CT, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| | - Magda Mocarska
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | | | | |
Collapse
|
12
|
Yoshioka Y, Taniguchi JB, Homma H, Tamura T, Fujita K, Inotsume M, Tagawa K, Misawa K, Matsumoto N, Nakagawa M, Inoue H, Tanaka H, Okazawa H. AAV-mediated editing of PMP22 rescues Charcot-Marie-Tooth disease type 1A features in patient-derived iPS Schwann cells. COMMUNICATIONS MEDICINE 2023; 3:170. [PMID: 38017287 PMCID: PMC10684506 DOI: 10.1038/s43856-023-00400-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Charcot-Marie-Tooth disease type 1A (CMT1A) is one of the most common hereditary peripheral neuropathies caused by duplication of 1.5 Mb genome region including PMP22 gene. We aimed to correct the duplication in human CMT1A patient-derived iPS cells (CMT1A-iPSCs) by genome editing and intended to analyze the effect on Schwann cells differentiated from CMT1A-iPSCs. METHODS We designed multiple gRNAs targeting a unique sequence present at two sites that sandwich only a single copy of duplicated peripheral myelin protein 22 (PMP22) genes, and selected one of them (gRNA3) from screening their efficiencies by T7E1 mismatch detection assay. AAV2-hSaCas9-gRNAedit was generated by subcloning gRNA3 into pX601-AAV-CMV plasmid, and the genome editing AAV vector was infected to CMT1A-iPSCs or CMT1A-iPSC-derived Schwann cell precursors. The effect of the genome editing AAV vector on myelination was evaluated by co-immunostaining of myelin basic protein (MBP), a marker of mature myelin, and microtubule-associated protein 2(MAP2), a marker of neurites or by electron microscopy. RESULTS Here we show that infection of CMT1A-iPS cells (iPSCs) with AAV2-hSaCas9-gRNAedit expressing both hSaCas9 and gRNA targeting the tandem repeat sequence decreased PMP22 gene duplication by 20-40%. Infection of CMT1A-iPSC-derived Schwann cell precursors with AAV2-hSaCas9-gRNAedit normalized PMP22 mRNA and PMP22 protein expression levels, and also ameliorated increased apoptosis and impaired myelination in CMT1A-iPSC-derived Schwann cells. CONCLUSIONS In vivo transfer of AAV2-hSaCas9-gRNAedit to peripheral nerves could be a potential therapeutic modality for CMT1A patient after careful examinations of toxicity including off-target mutations.
Collapse
Affiliation(s)
- Yuki Yoshioka
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Juliana Bosso Taniguchi
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hidenori Homma
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takuya Tamura
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kyota Fujita
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Maiko Inotsume
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kazuhiko Tagawa
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kazuharu Misawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
- RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Masanori Nakagawa
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, 606-8507, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
- Drug-discovery cellular basis development team, RIKEN BioResource Center, Kyoto, 606-8507, Japan
| | - Hikari Tanaka
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
13
|
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
|
14
|
Cavalcanti EBU, Leal RDCC, Marques Junior W, Nascimento OJMD. Charcot-Marie-Tooth disease: from historical landmarks in Brazil to current care perspectives. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:913-921. [PMID: 37611635 PMCID: PMC10631856 DOI: 10.1055/s-0043-1770348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/16/2023] [Indexed: 08/25/2023]
Abstract
Hereditary motor and sensory neuropathy, also known as Charcot-Marie-Tooth disease (CMT), traditionally refers to a group of genetic disorders in which neuropathy is the main or sole feature. Its prevalence varies according to different populations studied, with an estimate between 1:2,500 to 1:10,000. Since the identification of PMP22 gene duplication on chromosome 17 by Vance et al., in 1989, more than 100 genes have been related to this group of disorders, and we have seen advances in the care of patients, with identification of associated conditions and better supportive treatments, including clinical and surgical interventions. Also, with discoveries in the field of genetics, including RNA interference and gene editing techniques, new treatment perspectives begin to emerge. In the present work, we report the most import landmarks regarding CMT research in Brazil and provide a comprehensive review on topics such as frequency of different genes associated with CMT in our population, prevalence of pain, impact on pregnancy, respiratory features, and development of new therapies.
Collapse
Affiliation(s)
| | | | - Wilson Marques Junior
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Neurologia, Ribeirão Preto SP, Brazil.
| | | |
Collapse
|
15
|
Nair MA, Niu Z, Madigan NN, Shin AY, Brault JS, Staff NP, Klein CJ. Clinical trials in Charcot-Marie-Tooth disorders: a retrospective and preclinical assessment. Front Neurol 2023; 14:1251885. [PMID: 37808507 PMCID: PMC10556688 DOI: 10.3389/fneur.2023.1251885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Objective This study aimed to evaluate the progression of clinical and preclinical trials in Charcot-Marie-Tooth (CMT) disorders. Background CMT has historically been managed symptomatically and with genetic counseling. The evolution of molecular and pathologic understanding holds a therapeutic promise in gene-targeted therapies. Methods ClinicalTrials.gov from December 1999 to June 2022 was data extracted for CMT with preclinical animal gene therapy trials also reviewed by PubMed search. Results The number of active trials was 1 in 1999 and 286 in 2022. Academic settings accounted for 91% and pharmaceutical companies 9%. Of the pharmaceutical and academic trials, 38% and 28%, respectively, were controlled, randomized, and double-blinded. Thirty-two countries participated: the United States accounted for 26% (75/286). In total, 86% of the trials were classified as therapeutic: 50% procedural (21% wrist/elbow surgery; 22% shock wave and hydrodissection therapy), 23% investigational drugs, 15% devices, and 11% physical therapy. Sixty-seven therapeutic trials (49%) were designated phases 1-2 and 51% phases 3-4. The remaining 14% represent non-therapeutic trials: diagnostic testing (3%), functional outcomes (4%), natural history (4%), and standard of care (3%). One-hundred and three (36%) resulted in publications. Phase I human pharmaceutical trials are focusing on the safety of small molecule therapies (n = 8) and AAV and non-viral gene therapy (n = 3). Preclinical animal gene therapy studies include 11 different CMT forms including viral, CRISPR-Cas9, and nanoparticle delivery. Conclusion Current CMT trials are exploring procedural and molecular therapeutic options with substantial participation of the pharmaceutical industry worldwide. Emerging drug therapies directed at molecular pathogenesis are being advanced in human clinical trials; however, the majority remain within animal investigations.
Collapse
Affiliation(s)
- Malavika A. Nair
- Department of Graduate Education, Alix School of Medicine, Rochester, MN, United States
| | - Zhiyv Niu
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States
- Department of Clinical Genomics, Rochester, MN, United States
| | | | - Alexander Y. Shin
- Division of Hand Surgery, Department of Orthopaedic, Rochester, MN, United States
| | - Jeffrey S. Brault
- Department of Physical Medicine and Rehabilitation Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - Christopher J. Klein
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States
- Department of Neurology, Rochester, MN, United States
| |
Collapse
|
16
|
Wu TT, Finkel RS, Siskind CE, Feely SME, Burns J, Reilly MM, Muntoni F, Milev E, Estilow T, Shy ME, Ramchandren S. Validation of the parent-proxy version of the pediatric Charcot-Marie-Tooth disease quality of life instrument for children aged 0-7 years. J Peripher Nerv Syst 2023; 28:382-389. [PMID: 37166413 DOI: 10.1111/jns.12557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023]
Abstract
OBJECTIVE To evaluate the parent-proxy version of the pediatric Charcot Marie Tooth specific quality of life (pCMT-QOL) outcome instrument for children aged 7 or younger with CMT. We have previously developed and validated the direct-report pCMT-QOL for children aged 8-18 years and a parent proxy version of the instrument for children 8-18 years old. There is currently no CMT-QOL outcome measure for children aged 0-7 years old. METHODS Testing was conducted in parents or caregivers of children aged 0-7 years old with CMT evaluated at participating INC sites from the USA, United Kingdom, and Australia. The development of the instrument was iterative, involving identification of relevant domains, item pool generation, prospective pilot testing and clinical assessments, structured focus group interviews, and psychometric testing. The parent-proxy instrument was validated rigorously by examining previously identified domains and undergoing psychometric tests for children aged 0-7. RESULTS The parent-proxy pCMT-QOL working versions were administered to 128 parents/caregivers of children aged 0-7 years old between 2010 and 2016. The resulting data underwent rigorous psychometric analysis, including factor analysis, internal consistency, and convergent validity, and longitudinal analysis to develop the final parent-proxy version of the pCMT-QOL outcome measure for children aged 0-7 years old. CONCLUSIONS The parent-proxy version of the pCMT-QOL outcome measure, known as the pCMT-QOL (0-7 years parent-proxy) is a valid and sensitive proxy measure of health-related QOL for children aged 0-7 years with CMT.
Collapse
Affiliation(s)
- Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Carly E Siskind
- Department of Neurology, Stanford University, Stanford, California, USA
| | - Shawna M E Feely
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Joshua Burns
- Faculty of Medicine and Health, University of Sydney School of Health Sciences, Sydney, New South Wales, Australia
- Pediatric Gait Analysis Service of New South Wales, Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Francesco Muntoni
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Evelin Milev
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Timothy Estilow
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael E Shy
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Sindhu Ramchandren
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, Wayne State University, Detroit, Michigan, USA
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Titusville, New Jersey, USA
| |
Collapse
|
17
|
Pisciotta C, Pareyson D. Gene therapy and other novel treatment approaches for Charcot-Marie-Tooth disease. Neuromuscul Disord 2023; 33:627-635. [PMID: 37455204 DOI: 10.1016/j.nmd.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
There is still no effective drug treatment available for Charcot-Marie-Tooth disease (CMT). Current management relies on rehabilitation therapy, surgery for skeletal deformities, and symptomatic treatment. The challenge is to find disease-modifying therapies. Several approaches, including gene silencing (by means of ASO, siRNA, shRNA, miRNA, CRISPR-Cas9 editing), to counteract the PMP22 gene overexpression in the most frequent CMT1A type are under investigation. PXT3003 is the compound in the most advanced phase for CMT1A, as a second phase-III trial is ongoing. Gene therapy to substitute defective genes (particularly in recessive forms associated with loss-of-function mutations) or insert novel ones (e.g., NT3 gene) are being developed and tested in animal models and in still exceptional cases have reached the clinical trial phase in humans. Novel treatment approaches are also aimed at developing compounds acting on pathways important for different CMT types. Modulation of the neuregulin pathway determining myelin thickness is promising for both hypo-demyelinating and hypermyelinating neuropathies; intervention on Unfolded Protein Response seems effective for rescuing misfolded myelin proteins such as MPZ in CMT1B. HDAC6 inhibitors improved axonal transport and ameliorated phenotypes in different CMT models. Other potential therapeutic strategies include targeting macrophages, lipid metabolism, and Nav1.8 sodium channel in demyelinating CMT and the P2×7 receptor, which regulates calcium influx into Schwann cells, in CMT1A. Further approaches are aimed at correcting metabolic abnormalities, including the accumulation of sorbitol caused by biallelic mutations in the sorbitol dehydrogenase (SORD) gene and of neurotoxic glycosphingolipids in HSN1.
Collapse
Affiliation(s)
- Chiara Pisciotta
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Unit of Rare Neurological Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
18
|
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: 3.0] [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
|
19
|
Okamoto Y, Takashima H. The Current State of Charcot-Marie-Tooth Disease Treatment. Genes (Basel) 2023; 14:1391. [PMID: 37510296 PMCID: PMC10379063 DOI: 10.3390/genes14071391] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Charcot-Marie-Tooth disease (CMT) and associated neuropathies are the most predominant genetically transmitted neuromuscular conditions; however, effective pharmacological treatments have not established. The extensive genetic heterogeneity of CMT, which impacts the peripheral nerves and causes lifelong disability, presents a significant barrier to the development of comprehensive treatments. An estimated 100 loci within the human genome are linked to various forms of CMT and its related inherited neuropathies. This review delves into prospective therapeutic strategies used for the most frequently encountered CMT variants, namely CMT1A, CMT1B, CMTX1, and CMT2A. Compounds such as PXT3003, which are being clinically and preclinically investigated, and a broad array of therapeutic agents and their corresponding mechanisms are discussed. Furthermore, the progress in established gene therapy techniques, including gene replacement via viral vectors, exon skipping using antisense oligonucleotides, splicing modification, and gene knockdown, are appraised. Each of these gene therapies has the potential for substantial advancements in future research.
Collapse
Affiliation(s)
- Yuji Okamoto
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima 890-8544, Japan
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| |
Collapse
|
20
|
Stavrou M, Kleopa KA. CMT1A current gene therapy approaches and promising biomarkers. Neural Regen Res 2023; 18:1434-1440. [PMID: 36571339 PMCID: PMC10075121 DOI: 10.4103/1673-5374.361538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Charcot-Marie-Tooth neuropathies (CMT) constitute a group of common but highly heterogeneous, non-syndromic genetic disorders affecting predominantly the peripheral nervous system. CMT type 1A (CMT1A) is the most frequent type and accounts for almost ~50% of all diagnosed CMT cases. CMT1A results from the duplication of the peripheral myelin protein 22 (PMP22) gene. Overexpression of PMP22 protein overloads the protein folding apparatus in Schwann cells and activates the unfolded protein response. This leads to Schwann cell apoptosis, dys- and de- myelination and secondary axonal degeneration, ultimately causing neurological disabilities. During the last decades, several different gene therapies have been developed to treat CMT1A. Almost all of them remain at the pre-clinical stage using CMT1A animal models overexpressing PMP22. The therapeutic goal is to achieve gene silencing, directly or indirectly, thereby reversing the CMT1A genetic mechanism allowing the recovery of myelination and prevention of axonal loss. As promising treatments are rapidly emerging, treatment-responsive and clinically relevant biomarkers are becoming necessary. These biomarkers and sensitive clinical evaluation tools will facilitate the design and successful completion of future clinical trials for CMT1A.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics; Center for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
21
|
Wu TT, Finkel RS, Siskind CE, Feely SM, Burns J, Reilly MM, Muntoni F, Estilow T, Shy ME, Ramchandren S. Validation of the parent-proxy pediatric Charcot-Marie-Tooth disease quality of life outcome measure. J Peripher Nerv Syst 2023; 28:237-251. [PMID: 36748295 PMCID: PMC10521146 DOI: 10.1111/jns.12538] [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: 12/08/2022] [Revised: 01/16/2023] [Accepted: 01/28/2023] [Indexed: 02/08/2023]
Abstract
Charcot-Marie-Tooth disease (CMT) reduces health-related quality of life (QOL) in children. We have previously developed and validated the English and Italian versions of the pediatric CMT-specific QOL outcome measure (pCMT-QOL) for children aged 8 to 18. There is currently no parent-proxy CMT QOL outcome measure for use in clinical trials, which could provide complementary information in these children and adolescents. This study describes the validation studies conducted to develop the parent-proxy version of the pCMT-QOL outcome measure for children aged 8 to 18 years old. Development and validation of the parent-proxy version of the pCMT-QOL outcome measure for children aged 8 to 18 years old was iterative, involving identifying relevant domains, item pool generation, prospective pilot testing and clinical assessments, structured focus-group interviews, and psychometric testing, conducted on parents of children with CMT seen at participating sites from the USA, United Kingdom, and Australia. We utilized previously described methods to develop a working parent-proxy version of the pCMT-QOL measure. From 2010 to 2016, the parent-proxy pCMT-QOL working version was administered to 358 parents of children with CMT aged 8 to 18, seen at the participating study sites of the Inherited Neuropathies Consortium. The resulting data underwent rigorous psychometric analysis, including factor analysis, test-retest reliability, internal consistency, convergent validity, IRT analysis, and longitudinal analysis, to develop the final parent-proxy version of the pCMT-QOL outcome measure for children aged 8 to 18 years old. The parent-proxy version of the pCMT-QOL outcome measure is a reliable, valid, and sensitive proxy measure of health-related QOL for children aged 8 to 18 with CMT.
Collapse
Affiliation(s)
- Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Richard S. Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Shawna M.E. Feely
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Joshua Burns
- University of Sydney School of Health Sciences, Faculty of Medicine and Health; Pediatric Gait Analysis Service of New South Wales, Sydney Children’s Hospitals Network, Sydney, Australia
| | - Mary M. Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Francesco Muntoni
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Timothy Estilow
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael E. Shy
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sindhu Ramchandren
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
- The Janssen Pharmaceutical Companies of Johnson & Johnson, NJ, USA
| | | |
Collapse
|
22
|
Putka AF, Mato JP, McLoughlin HS. Myelinating Glia: Potential Therapeutic Targets in Polyglutamine Spinocerebellar Ataxias. Cells 2023; 12:601. [PMID: 36831268 PMCID: PMC9953858 DOI: 10.3390/cells12040601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Human studies, in combination with animal and cellular models, support glial cells as both major contributors to neurodegenerative diseases and promising therapeutic targets. Among glial cells, oligodendrocytes and Schwann cells are the myelinating glial cells of the central and peripheral nervous system, respectively. In this review, we discuss the contributions of these central and peripheral myelinating glia to the pathomechanisms of polyglutamine (polyQ) spinocerebellar ataxia (SCA) types 1, 2, 3, 6, 7, and 17. First, we highlight the function of oligodendrocytes in healthy conditions and how they are disrupted in polyQ SCA patients and diseased model systems. We then cover the role of Schwann cells in peripheral nerve function and repair as well as their possible role in peripheral neuropathy in polyQ SCAs. Finally, we discuss potential polyQ SCA therapeutic interventions in myelinating glial.
Collapse
Affiliation(s)
- Alexandra F. Putka
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Juan P. Mato
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
23
|
Malong L, Napoli I, Casal G, White IJ, Stierli S, Vaughan A, Cattin AL, Burden JJ, Hng KI, Bossio A, Flanagan A, Zhao HT, Lloyd AC. Characterization of the structure and control of the blood-nerve barrier identifies avenues for therapeutic delivery. Dev Cell 2023; 58:174-191.e8. [PMID: 36706755 DOI: 10.1016/j.devcel.2023.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 10/26/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
The blood barriers of the nervous system protect neural environments but can hinder therapeutic accessibility. The blood-brain barrier (BBB) is well characterized, consisting of endothelial cells with specialized tight junctions and low levels of transcytosis, properties conferred by contacting pericytes and astrocytes. In contrast, the blood-nerve barrier (BNB) of the peripheral nervous system is poorly defined. Here, we characterize the structure of the mammalian BNB, identify the processes that confer barrier function, and demonstrate how the barrier can be opened in response to injury. The homeostatic BNB is leakier than the BBB, which we show is due to higher levels of transcytosis. However, the barrier is reinforced by macrophages that specifically engulf leaked materials, identifying a role for resident macrophages as an important component of the BNB. Finally, we demonstrate the exploitation of these processes to effectively deliver RNA-targeting therapeutics to peripheral nerves, indicating new treatment approaches for nervous system pathologies.
Collapse
Affiliation(s)
- Liza Malong
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ilaria Napoli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Giulia Casal
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ian J White
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Salome Stierli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Andrew Vaughan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jemima J Burden
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Keng I Hng
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Alessandro Bossio
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Adrienne Flanagan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Hien T Zhao
- IONIS, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
24
|
Saidia AR, Ruel J, Bahloul A, Chaix B, Venail F, Wang J. Current Advances in Gene Therapies of Genetic Auditory Neuropathy Spectrum Disorder. J Clin Med 2023; 12:jcm12030738. [PMID: 36769387 PMCID: PMC9918155 DOI: 10.3390/jcm12030738] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) refers to a range of hearing impairments characterized by an impaired transmission of sound from the cochlea to the brain. This defect can be due to a lesion or defect in the inner hair cell (IHC), IHC ribbon synapse (e.g., pre-synaptic release of glutamate), postsynaptic terminals of the spiral ganglion neurons, or demyelination and axonal loss within the auditory nerve. To date, the only clinical treatment options for ANSD are hearing aids and cochlear implantation. However, despite the advances in hearing-aid and cochlear-implant technologies, the quality of perceived sound still cannot match that of the normal ear. Recent advanced genetic diagnostics and clinical audiology made it possible to identify the precise site of a lesion and to characterize the specific disease mechanisms of ANSD, thus bringing renewed hope to the treatment or prevention of auditory neurodegeneration. Moreover, genetic routes involving the replacement or corrective editing of mutant sequences or defected genes to repair damaged cells for the future restoration of hearing in deaf people are showing promise. In this review, we provide an update on recent discoveries in the molecular pathophysiology of genetic lesions, auditory synaptopathy and neuropathy, and gene-therapy research towards hearing restoration in rodent models and in clinical trials.
Collapse
Affiliation(s)
- Anissa Rym Saidia
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, 34295 Montpellier, France
| | - Jérôme Ruel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, 34295 Montpellier, France
- Cognitive Neuroscience Laboratory, Aix-Marseille University, CNRS, UMR 7291, 13331 Marseille, France
| | - Amel Bahloul
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, 34295 Montpellier, France
| | - Benjamin Chaix
- Department of ENT and Head and Neck Surgery, University Hospital of Montpellier, 34295 Montpellier, France
| | - Frédéric Venail
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, 34295 Montpellier, France
- Department of ENT and Head and Neck Surgery, University Hospital of Montpellier, 34295 Montpellier, France
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, 34295 Montpellier, France
- Department of ENT and Head and Neck Surgery, University Hospital of Montpellier, 34295 Montpellier, France
- Correspondence: ; Tel.: +33-499-63-60-48
| |
Collapse
|
25
|
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: 2] [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
|
26
|
Therapeutic tools for inherited neuropathies. Rev Neurol (Paris) 2023; 179:5-9. [PMID: 36529569 DOI: 10.1016/j.neurol.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inherited neuropathies are a genetically and phenotypically heterogenous group of disorders leading to sensory and motor dysfunction. For years, these neuropathies have been considered as non-treatable diseases, as no drug is able to induce nerve regrowth. Progress in molecular tools has changed this view and several neuropathies can now be efficiently treated. Some more will be treatable in the upcoming years. Basically, these new treatments can be divided into four categories, depending on the target: gene therapy; gene expression therapy; protein modification or replacement (enzyme replacement therapy, ERT); downstream therapies. In this short review, we will provide a few examples for each of them in the field of peripheral neuropathies.
Collapse
|
27
|
Beloribi-Djefaflia S, Attarian S. Treatment of Charcot-Marie-Tooth neuropathies. Rev Neurol (Paris) 2023; 179:35-48. [PMID: 36588067 DOI: 10.1016/j.neurol.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022]
Abstract
Charcot-Marie-Tooth (CMT) is a heterogeneous group of inherited neuropathies that affect the peripheral nerves and slowly cause progressive disability. Currently, there is no effective therapy. Patients' management is based on rehabilitation and occupational therapy, fatigue, and pain treatment with regular follow-up according to the severity of the disease. In the last three decades, much progress has been made to identify mutations involved in the different types of CMT, decipher the pathophysiology of the disease, and identify key genes and pathways that could be targeted to propose new therapeutic strategies. Genetic therapy is one of the fields of interest to silence genes such as PMP22 in CMT1A or to express GJB1 in CMT1X. Among the most promising molecules, inhibitors of the NRG-1 axis and modulators of UPR or the HDACs enzyme family could be used in different types of CMT.
Collapse
Affiliation(s)
- S Beloribi-Djefaflia
- Reference center for neuromuscular disorders and ALS, AP-HM, CHU La Timone, Marseille, France
| | - S Attarian
- Reference center for neuromuscular disorders and ALS, AP-HM, CHU La Timone, Marseille, France; FILNEMUS, European Reference Network for Rare Diseases (ERN), Marseille, France; Medical Genetics, Aix Marseille Université-Inserm UMR_1251, 13005 Marseille, France.
| |
Collapse
|
28
|
Younger DS. On the path to evidence-based therapy in neuromuscular disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:315-358. [PMID: 37562877 DOI: 10.1016/b978-0-323-98818-6.00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Neuromuscular disorders encompass a diverse group of acquired and genetic diseases characterized by loss of motor functionality. Although cure is the goal, many therapeutic strategies have been envisioned and are being studied in randomized clinical trials and entered clinical practice. As in all scientific endeavors, the successful clinical translation depends on the quality and translatability of preclinical findings and on the predictive value and feasibility of the clinical models. This chapter focuses on five exemplary diseases: childhood spinal muscular atrophy (SMA), Charcot-Marie-Tooth (CMT) disorders, chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), acquired autoimmune myasthenia gravis (MG), and Duchenne muscular dystrophy (DMD), to illustrate the progress made on the path to evidenced-based therapy.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
29
|
Pisciotta C, Shy ME. Hereditary neuropathy. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:609-617. [PMID: 37562889 DOI: 10.1016/b978-0-323-98818-6.00009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The hereditary neuropathies, collectively referred as Charcot-Marie-Tooth disease (CMT) and related disorders, are heterogeneous genetic peripheral nerve disorders that collectively comprise the commonest inherited neurological disease with an estimated prevalence of 1:2500 individuals. The field of hereditary neuropathies has made significant progress in recent years with respect to both gene discovery and treatment as a result of next-generation sequencing (NGS) approach. These investigations which have identified over 100 causative genes and new mutations have made the classification of CMT even more challenging. Despite so many different mutated genes, the majority of CMT forms share a similar clinical phenotype, and due to this phenotypic homogeneity, genetic testing in CMT is increasingly being performed through the use of NGS panels. The majority of patients still have a mutation in one the four most common genes (PMP22 duplication-CMT1A, MPZ-CMT1B, GJB1-CMTX1, and MFN2-CMT2A). This chapter focuses primarily on these four forms and their potential therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Pisciotta
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Michael E Shy
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
30
|
Amanat M, Nemeth CL, Fine AS, Leung DG, Fatemi A. Antisense Oligonucleotide Therapy for the Nervous System: From Bench to Bedside with Emphasis on Pediatric Neurology. Pharmaceutics 2022; 14:2389. [PMID: 36365206 PMCID: PMC9695718 DOI: 10.3390/pharmaceutics14112389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 09/05/2023] Open
Abstract
Antisense oligonucleotides (ASOs) are disease-modifying agents affecting protein-coding and noncoding ribonucleic acids. Depending on the chemical modification and the location of hybridization, ASOs are able to reduce the level of toxic proteins, increase the level of functional protein, or modify the structure of impaired protein to improve function. There are multiple challenges in delivering ASOs to their site of action. Chemical modifications in the phosphodiester bond, nucleotide sugar, and nucleobase can increase structural thermodynamic stability and prevent ASO degradation. Furthermore, different particles, including viral vectors, conjugated peptides, conjugated antibodies, and nanocarriers, may improve ASO delivery. To date, six ASOs have been approved by the US Food and Drug Administration (FDA) in three neurological disorders: spinal muscular atrophy, Duchenne muscular dystrophy, and polyneuropathy caused by hereditary transthyretin amyloidosis. Ongoing preclinical and clinical studies are assessing the safety and efficacy of ASOs in multiple genetic and acquired neurological conditions. The current review provides an update on underlying mechanisms, design, chemical modifications, and delivery of ASOs. The administration of FDA-approved ASOs in neurological disorders is described, and current evidence on the safety and efficacy of ASOs in other neurological conditions, including pediatric neurological disorders, is reviewed.
Collapse
Affiliation(s)
- Man Amanat
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christina L. Nemeth
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amena Smith Fine
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Doris G. Leung
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Genetic Muscle Disorders, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
31
|
Intisar A, Shin HY, Kim W, Kang HG, Kim MY, Kim YS, Cho Y, Mo YJ, Lim H, Lee S, Lu QR, Lee Y, Kim MS. Implantable Electroceutical Approach Improves Myelination by Restoring Membrane Integrity in a Mouse Model of Peripheral Demyelinating Neuropathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201358. [PMID: 35975427 PMCID: PMC9661852 DOI: 10.1002/advs.202201358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Although many efforts are undertaken to treat peripheral demyelinating neuropathies based on biochemical interventions, unfortunately, there is no approved treatment yet. Furthermore, previous studies have not shown improvement of the myelin membrane at the biomolecular level. Here, an electroceutical treatment is introduced as a biophysical intervention to treat Charcot-Marie-Tooth (CMT) disease-the most prevalent peripheral demyelinating neuropathy worldwide-using a mouse model. The specific electrical stimulation (ES) condition (50 mV mm-1 , 20 Hz, 1 h) for optimal myelination is found via an in vitro ES screening system, and its promyelinating effect is validated with ex vivo dorsal root ganglion model. Biomolecular investigation via time-of-flight secondary ion mass spectrometry shows that ES ameliorates distribution abnormalities of peripheral myelin protein 22 and cholesterol in the myelin membrane, revealing the restoration of myelin membrane integrity. ES intervention in vivo via flexible implantable electrodes shows not only gradual rehabilitation of mouse behavioral phenotypes (balance and endurance), but also restored myelin thickness, compactness, and membrane integrity. This study demonstrates, for the first time, that an electroceutical approach with the optimal ES condition has the potential to treat CMT disease and restore impaired myelin membrane integrity, shifting the paradigm toward practical interventions for peripheral demyelinating neuropathies.
Collapse
Affiliation(s)
- Aseer Intisar
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Hyun Young Shin
- CTCELLS Corp.Daegu42988Republic of Korea
- SBCure Corp.Daegu43017Republic of Korea
| | | | - Hyun Gyu Kang
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Min Young Kim
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Yu Seon Kim
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Youngjun Cho
- Department of Robotics and Mechatronics EngineeringDGISTDaegu42988Republic of Korea
| | - Yun Jeoung Mo
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Heejin Lim
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Sanghoon Lee
- Department of Robotics and Mechatronics EngineeringDGISTDaegu42988Republic of Korea
| | - Q. Richard Lu
- Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
| | - Yun‐Il Lee
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Minseok S. Kim
- Department of New BiologyDGISTDaegu42988Republic of Korea
- CTCELLS Corp.Daegu42988Republic of Korea
- Translational Responsive Medicine Center (TRMC)DGISTDaegu42988Republic of Korea
- New Biology Research Center (NBRC)DGISTDaegu42988Republic of Korea
| |
Collapse
|
32
|
Nguyen NP, Lopez S, Smith CL, Lever TE, Nichols NL, Bunyak F. Axon and Myelin Sheath Segmentation in Electron Microscopy Images using Meta Learning. IEEE APPLIED IMAGERY PATTERN RECOGNITION WORKSHOP : [PROCEEDINGS]. IEEE APPLIED IMAGERY PATTERN RECOGNITION WORKSHOP 2022; 2022:10.1109/aipr57179.2022.10092238. [PMID: 37214276 PMCID: PMC10197949 DOI: 10.1109/aipr57179.2022.10092238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Various neurological diseases affect the morphology of myelinated axons. Quantitative analysis of these structures and changes occurring due to neurodegeneration or neuroregeneration is of great importance for characterization of disease state and treatment response. This paper proposes a robust, meta-learning based pipeline for segmentation of axons and surrounding myelin sheaths in electron microscopy images. This is the first step towards computation of electron microscopy related bio-markers of hypoglossal nerve degeneration/regeneration. This segmentation task is challenging due to large variations in morphology and texture of myelinated axons at different levels of degeneration and very limited availability of annotated data. To overcome these difficulties, the proposed pipeline uses a meta learning-based training strategy and a U-net like encoder decoder deep neural network. Experiments on unseen test data collected at different magnification levels (i.e, trained on 500X and 1200X images, and tested on 250X and 2500X images) showed improved segmentation performance by 5% to 7% compared to a regularly trained, comparable deep learning network.
Collapse
Affiliation(s)
- Nguyen P. Nguyen
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA
| | - Stephanie Lopez
- Department of Biomedical Sciences, Department of Otolaryngology-Head and Neck Surgery, University of Missouri-Columbia, MO, USA
| | - Catherine L. Smith
- Department of Biomedical Sciences, Department of Otolaryngology-Head and Neck Surgery, University of Missouri-Columbia, MO, USA
| | - Teresa E. Lever
- Department of Biomedical Sciences, Department of Otolaryngology-Head and Neck Surgery, University of Missouri-Columbia, MO, USA
| | - Nicole L. Nichols
- Department of Biomedical Sciences, Department of Otolaryngology-Head and Neck Surgery, University of Missouri-Columbia, MO, USA
| | - Filiz Bunyak
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA
| |
Collapse
|
33
|
Elbaz B, Yang L, Vardy M, Isaac S, Rader BL, Kawaguchi R, Traka M, Woolf CJ, Renthal W, Popko B. Sensory neurons display cell-type-specific vulnerability to loss of neuron-glia interactions. Cell Rep 2022; 40:111130. [PMID: 35858549 PMCID: PMC9354470 DOI: 10.1016/j.celrep.2022.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/22/2021] [Accepted: 07/01/2022] [Indexed: 11/11/2022] Open
Abstract
Peripheral nervous system (PNS) injuries initiate transcriptional changes in glial cells and sensory neurons that promote axonal regeneration. While the factors that initiate the transcriptional changes in glial cells are well characterized, the full range of stimuli that initiate the response of sensory neurons remain elusive. Here, using a genetic model of glial cell ablation, we find that glial cell loss results in transient PNS demyelination without overt axonal loss. By profiling sensory ganglia at single-cell resolution, we show that glial cell loss induces a transcriptional injury response preferentially in proprioceptive and Aβ RA-LTMR neurons. The transcriptional response of sensory neurons to mechanical injury has been assumed to be a cell-autonomous response. By identifying a similar response in non-injured, demyelinated neurons, our study suggests that this represents a non-cell-autonomous transcriptional response of sensory neurons to glial cell loss and demyelination.
Collapse
Affiliation(s)
- Benayahu Elbaz
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA.
| | - Lite Yang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Maia Vardy
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Sara Isaac
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Braesen L Rader
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maria Traka
- Department of Anatomy, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA 02115, USA
| | - William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Brian Popko
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
34
|
Polikarpova AV, Egorova TV, Bardina MV. Genetically modified animal models of hereditary diseases for testing of gene-directed therapy. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.82618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Disease-causing genes have been identified for many severe muscular and neurological genetic disorders. Advances in the gene therapy field offer promising solutions for drug development to treat these life-threatening conditions. Depending on how the mutation affects the function of the gene product, different gene therapy approaches may be beneficial. Gene replacement therapy is appropriate for diseases caused by mutations that result in the deficiency of the functional protein. Gene suppression strategy is suggested for disorders caused by the toxic product of the mutant gene. Splicing modulators, genome editing, and base editing techniques can be applied to disorders with different types of underlying mutations. Testing potential drugs in animal models of human diseases is an indispensable step of development. Given the specific gene therapy approach, appropriate animal models can be generated using a variety of technologies ranging from transgenesis to precise genome editing. In this review, we discuss technologies used to generate small and large animal models of the most common muscular and neurological genetic disorders. We specifically focus on animal models that were used to test gene therapies based on adeno-associated vectors and antisense nucleotides.
Collapse
|
35
|
Prior R, Verschoren S, Vints K, Jaspers T, Rossaert E, Klingl YE, Silva A, Hersmus N, Van Damme P, Van Den Bosch L. HDAC3 Inhibition Stimulates Myelination in a CMT1A Mouse Model. Mol Neurobiol 2022; 59:3414-3430. [PMID: 35320455 PMCID: PMC9148289 DOI: 10.1007/s12035-022-02782-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 02/07/2022] [Indexed: 12/02/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) is the most common inherited peripheral neuropathy, with currently no effective treatment or cure. CMT1A is caused by a duplication of the PMP22 gene, which leads to Schwann cell differentiation defects and dysmyelination of the peripheral nerves. The epigenetic regulator histone deacetylase 3 (HDAC3) has been shown to negatively regulate myelination as well as its associated signaling pathways, PI3K-AKT and MAPK-ERK. We showed that these signaling pathways are indeed downregulated in the C3-PMP22 mouse model, similar to what has been shown in the CMT1A rat model. We confirmed that early postnatal defects are present in the peripheral nerves of the C3-PMP22 mouse model, which led to a progressive reduction in axon caliber size and myelination. The aim of this study was to investigate whether pharmacological HDAC3 inhibition could be a valuable therapeutic approach for this CMT1A mouse model. We demonstrated that early treatment of CMT1A mice with the selective HDAC3 inhibitor RGFP966 increased myelination and myelin g-ratios, which was associated with improved electrophysiological recordings. However, a high dose of RGFP966 caused a decline in rotarod performance and a decline in overall grip strength. Additionally, macrophage presence in peripheral nerves was increased in RGFP966 treated CMT1A mice. We conclude that HDAC3 does not only play a role in regulating myelination but is also important in the neuroimmune modulation. Overall, our results indicate that correct dosing of HDAC3 inhibitors is of crucial importance if translated to a clinical setting for demyelinating forms of CMT or other neurological disorders.
Collapse
Affiliation(s)
- Robert Prior
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium.
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium.
| | - Stijn Verschoren
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Katlijn Vints
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Electron Microscopy Platform & VIB BioImaging Core, Herestraat 49, B-3000, Leuven, Belgium
| | - Tom Jaspers
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Elisabeth Rossaert
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Yvonne E Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Alessio Silva
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Nicole Hersmus
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium
- Neurology, University Hospitals Leuven, B-3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, B-3000, Leuven, Belgium.
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Campus Gasthuisberg O&N5, Herestraat 49, box 602, B-3000, Leuven, Belgium.
| |
Collapse
|
36
|
Kramarz C, Rossor AM. Neurological update: hereditary neuropathies. J Neurol 2022; 269:5187-5191. [PMID: 35596796 PMCID: PMC9363318 DOI: 10.1007/s00415-022-11164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
In this update, we review the recent discovery of autosomal recessive variants in sorbitol dehydrogenase as one of the commonest and potentially treatable causes of hereditary motor neuropathy and CMT2. We also report on recent therapeutic advances in hereditary neuropathy including the use of lipid nanoparticle sequestered antisense oligonucleotides in CMT1A and lipid nanoparticle delivered CRISPR-Cas9 gene editing in ATTR amyloidosis.
Collapse
Affiliation(s)
- Caroline Kramarz
- Department of Neuromuscular Disease, Queen Square UCL Institute of Neurology and the National Hospital of Neurology and Neurosurgery, London, WC1N 3BG, UK
| | - Alexander M Rossor
- Department of Neuromuscular Disease, Queen Square UCL Institute of Neurology and the National Hospital of Neurology and Neurosurgery, London, WC1N 3BG, UK.
| |
Collapse
|
37
|
Stavrou M, Kagiava A, Choudury SG, Jennings MJ, Wallace LM, Fowler AM, Heslegrave A, Richter J, Tryfonos C, Christodoulou C, Zetterberg H, Horvath R, Harper SQ, Kleopa KA. A translatable RNAi-driven gene therapy silences PMP22/Pmp22 genes and improves neuropathy in CMT1A mice. J Clin Invest 2022; 132:159814. [PMID: 35579942 PMCID: PMC9246392 DOI: 10.1172/jci159814] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A), the most common inherited demyelinating peripheral neuropathy, is caused by PMP22 gene duplication. Overexpression of WT PMP22 in Schwann cells destabilizes the myelin sheath, leading to demyelination and ultimately to secondary axonal loss and disability. No treatments currently exist that modify the disease course. The most direct route to CMT1A therapy will involve reducing PMP22 to normal levels. To accomplish this, we developed a gene therapy strategy to reduce PMP22 using artificial miRNAs targeting human PMP22 and mouse Pmp22 mRNAs. Our lead therapeutic miRNA, miR871, was packaged into an adeno-associated virus 9 (AAV9) vector and delivered by lumbar intrathecal injection into C61-het mice, a model of CMT1A. AAV9-miR871 efficiently transduced Schwann cells in C61-het peripheral nerves and reduced human and mouse PMP22 mRNA and protein levels. Treatment at early and late stages of the disease significantly improved multiple functional outcome measures and nerve conduction velocities. Furthermore, myelin pathology in lumbar roots and femoral motor nerves was ameliorated. The treated mice also showed reductions in circulating biomarkers of CMT1A. Taken together, our data demonstrate that AAV9-miR871–driven silencing of PMP22 rescues a CMT1A model and provides proof of principle for treating CMT1A using a translatable gene therapy approach.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Sarah G Choudury
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, United States of America
| | - Matthew J Jennings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Lindsay M Wallace
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, United States of America
| | - Allison M Fowler
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, United States of America
| | - Amanda Heslegrave
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Jan Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christina Tryfonos
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christina Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Henrik Zetterberg
- Institute of Laboratory Medicine, Göteborgs University, Göteborg, Sweden
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Scott Q Harper
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, United States of America
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
38
|
Canalis E, Carrer M, Eller T, Schilling L, Yu J. Use of antisense oligonucleotides to target Notch3 in skeletal cells. PLoS One 2022; 17:e0268225. [PMID: 35536858 PMCID: PMC9089911 DOI: 10.1371/journal.pone.0268225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/25/2022] [Indexed: 01/02/2023] Open
Abstract
Notch receptors are determinants of cell fate and function, and play an important role in the regulation of bone development and skeletal remodeling. Lateral Meningocele Syndrome (LMS) is a monogenic disorder associated with NOTCH3 pathogenic variants that result in the stabilization of NOTCH3 and a gain-of-function. LMS presents with neurological developmental abnormalities and bone loss. We created a mouse model (Notch3em1Ecan) harboring a 6691TAATGA mutation in the Notch3 locus, and heterozygous Notch3em1Ecan mice exhibit cancellous and cortical bone osteopenia. In the present work, we explored whether Notch3 antisense oligonucleotides (ASO) downregulate Notch3 and have the potential to ameliorate the osteopenia of Notch3em1Ecan mice. Notch3 ASOs decreased the expression of Notch3 wild type and Notch36691-TAATGA mutant mRNA expressed by Notch3em1Ecan mice in osteoblast cultures without evidence of cellular toxicity. The effect was specific since ASOs did not downregulate Notch1, Notch2 or Notch4. The expression of Notch3 wild type and Notch36691-TAATGA mutant transcripts also was decreased in bone marrow stromal cells and osteocytes following exposure to Notch3 ASOs. In vivo, the subcutaneous administration of Notch3 ASOs at 25 to 50 mg/Kg decreased Notch3 mRNA in the liver, heart and bone. Microcomputed tomography demonstrated that the administration of Notch3 ASOs ameliorates the cortical osteopenia of Notch3em1Ecan mice, and ASOs decreased femoral cortical porosity and increased cortical thickness and bone volume. However, the administration of Notch3 ASOs did not ameliorate the cancellous bone osteopenia of Notchem1Ecan mice. In conclusion, Notch3 ASOs downregulate Notch3 expression in skeletal cells and their systemic administration ameliorates cortical osteopenia in Notch3em1Ecan mice; as such ASOs may become useful strategies in the management of skeletal diseases affected by Notch gain-of-function.
Collapse
MESH Headings
- Abnormalities, Multiple
- Animals
- Bone Diseases, Metabolic/genetics
- Bone Diseases, Metabolic/metabolism
- Bone and Bones/diagnostic imaging
- Bone and Bones/metabolism
- Meningocele
- Mice
- Oligonucleotides, Antisense
- RNA, Messenger
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Receptors, Notch/genetics
- X-Ray Microtomography
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- Department of Medicine, UConn Health, Farmington, Connecticut, United States of America
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Michele Carrer
- Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Tabitha Eller
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Lauren Schilling
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| |
Collapse
|
39
|
Bai Y, Treins C, Volpi VG, Scapin C, Ferri C, Mastrangelo R, Touvier T, Florio F, Bianchi F, Del Carro U, Baas FF, Wang D, Miniou P, Guedat P, Shy ME, D'Antonio M. Treatment with IFB-088 Improves Neuropathy in CMT1A and CMT1B Mice. Mol Neurobiol 2022; 59:4159-4178. [PMID: 35501630 PMCID: PMC9167212 DOI: 10.1007/s12035-022-02838-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/09/2022] [Indexed: 11/24/2022]
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A), caused by duplication of the peripheral myelin protein 22 (PMP22) gene, and CMT1B, caused by mutations in myelin protein zero (MPZ) gene, are the two most common forms of demyelinating CMT (CMT1), and no treatments are available for either. Prior studies of the MpzSer63del mouse model of CMT1B have demonstrated that protein misfolding, endoplasmic reticulum (ER) retention and activation of the unfolded protein response (UPR) contributed to the neuropathy. Heterozygous patients with an arginine to cysteine mutation in MPZ (MPZR98C) develop a severe infantile form of CMT1B which is modelled by MpzR98C/ + mice that also show ER stress and an activated UPR. C3-PMP22 mice are considered to effectively model CMT1A. Altered proteostasis, ER stress and activation of the UPR have been demonstrated in mice carrying Pmp22 mutations. To determine whether enabling the ER stress/UPR and readjusting protein homeostasis would effectively treat these models of CMT1B and CMT1A, we administered Sephin1/IFB-088/icerguestat, a UPR modulator which showed efficacy in the MpzS63del model of CMT1B, to heterozygous MpzR98C and C3-PMP22 mice. Mice were analysed by behavioural, neurophysiological, morphological and biochemical measures. Both MpzR98C/ + and C3-PMP22 mice improved in motor function and neurophysiology. Myelination, as demonstrated by g-ratios and myelin thickness, improved in CMT1B and CMT1A mice and markers of UPR activation returned towards wild-type values. Taken together, our results demonstrate the capability of IFB-088 to treat a second mouse model of CMT1B and a mouse model of CMT1A, the most common form of CMT. Given the recent benefits of IFB-088 treatment in amyotrophic lateral sclerosis and multiple sclerosis animal models, these data demonstrate its potential in managing UPR and ER stress for multiple mutations in CMT1 as well as in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yunhong Bai
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Vera G Volpi
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Cristina Scapin
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Cinzia Ferri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Rosa Mastrangelo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Thierry Touvier
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Francesca Florio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Francesca Bianchi
- Division of Neuroscience, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy
| | - Frank F Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - David Wang
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | | | | | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Maurizio D'Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute DIBIT, 20132, Milan, Italy.
| |
Collapse
|
40
|
Klein D, Groh J, Yuan X, Berve K, Stassart R, Fledrich R, Martini R. Early targeting of endoneurial macrophages alleviates the neuropathy and affects abnormal Schwann cell differentiation in a mouse model of Charcot-Marie-Tooth 1A. Glia 2022; 70:1100-1116. [PMID: 35188681 DOI: 10.1002/glia.24158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022]
Abstract
We have previously shown that targeting endoneurial macrophages with the orally applied CSF-1 receptor specific kinase (c-FMS) inhibitor PLX5622 from the age of 3 months onwards led to a substantial alleviation of the neuropathy in mouse models of Charcot-Marie-Tooth (CMT) 1X and 1B disease, which are genetically-mediated nerve disorders not treatable in humans. The same approach failed in a model of CMT1A (PMP22-overexpressing mice, line C61), representing the most frequent form of CMT. This was unexpected since previous studies identified macrophages contributing to disease severity in the same CMT1A model. Here we re-approached the possibility of alleviating the neuropathy in a model of CMT1A by targeting macrophages at earlier time points. As a proof-of-principle experiment, we genetically inactivated colony-stimulating factor-1 (CSF-1) in CMT1A mice, which resulted in lower endoneurial macrophage numbers and alleviated the neuropathy. Based on these observations, we pharmacologically ablated macrophages in newborn CMT1A mice by feeding their lactating mothers with chow containing PLX5622, followed by treatment of the respective progenies after weaning until the age of 6 months. We found that peripheral neuropathy was substantially alleviated after early postnatal treatment, leading to preserved motor function in CMT1A mice. Moreover, macrophage depletion affected the altered Schwann cell differentiation phenotype. These findings underscore the targetable role of macrophage-mediated inflammation in peripheral nerves of inherited neuropathies, but also emphasize the need for an early treatment start confined to a narrow therapeutic time window in CMT1A models and potentially in respective patients.
Collapse
Affiliation(s)
- Dennis Klein
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Xidi Yuan
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Kristina Berve
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Ruth Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Robert Fledrich
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
41
|
Motley W, Chaudry V, Lloyd TE. Treatment and Management of Hereditary Neuropathies. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
42
|
Pisciotta C, Saveri P, Pareyson D. Challenges in Treating Charcot-Marie-Tooth Disease and Related Neuropathies: Current Management and Future Perspectives. Brain Sci 2021; 11:1447. [PMID: 34827446 PMCID: PMC8615778 DOI: 10.3390/brainsci11111447] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/06/2023] Open
Abstract
There is still no effective drug treatment available for Charcot-Marie-Tooth neuropathies (CMT). Current management relies on rehabilitation therapy, surgery for skeletal deformities, and symptomatic treatment of pain; fatigue and cramps are frequent complaints that are difficult to treat. The challenge is to find disease-modifying therapies. Several approaches, including gene silencing, to counteract the PMP22 gene overexpression in the most frequent CMT1A type are under investigation. PXT3003 is the compound in the most advanced phase for CMT1A, as a second-phase III trial is ongoing. Gene therapy to substitute defective genes or insert novel ones and compounds acting on pathways important for different CMT types are being developed and tested in animal models. Modulation of the Neuregulin pathway determining myelin thickness is promising for both hypo-demyelinating and hypermyelinating neuropathies; intervention on Unfolded Protein Response seems effective for rescuing misfolded myelin proteins such as P0 in CMT1B. HDAC6 inhibitors improved axonal transport and ameliorated phenotypes in different CMT models. Other potential therapeutic strategies include targeting macrophages, lipid metabolism, and Nav1.8 sodium channel in demyelinating CMT and the P2X7 receptor, which regulates calcium influx into Schwann cells, in CMT1A. Further approaches are aimed at correcting metabolic abnormalities, including the accumulation of sorbitol caused by biallelic mutations in the sorbitol dehydrogenase (SORD) gene and of neurotoxic glycosphingolipids in HSN1.
Collapse
Affiliation(s)
| | | | - Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.P.); (P.S.)
| |
Collapse
|
43
|
Abstract
Demyelinating forms of Charcot-Marie-Tooth disease (CMT) are genetically and phenotypically heterogeneous and result from highly diverse biological mechanisms including gain of function (including dominant negative effects) and loss of function. While no definitive treatment is currently available, rapid advances in defining the pathomechanisms of demyelinating CMT have led to promising pre-clinical studies, as well as emerging clinical trials. Especially promising are the recently completed pre-clinical genetic therapy studies in PMP-22, GJB1, and SH3TC2-associated neuropathies, particularly given the success of similar approaches in humans with spinal muscular atrophy and transthyretin familial polyneuropathy. This article focuses on neuropathies related to mutations in PMP-22, MPZ, and GJB1, which together comprise the most common forms of demyelinating CMT, as well as on select rarer forms for which promising treatment targets have been identified. Clinical characteristics and pathomechanisms are reviewed in detail, with emphasis on therapeutically targetable biological pathways. Also discussed are the challenges facing the CMT research community in its efforts to advance the rapidly evolving biological insights to effective clinical trials. These considerations include the limitations of currently available animal models, the need for personalized medicine approaches/allele-specific interventions for select forms of demyelinating CMT, and the increasing demand for optimal clinical outcome assessments and objective biomarkers.
Collapse
Affiliation(s)
- Vera Fridman
- Department of Neurology, University of Colorado Anschutz Medical Campus, 12631 E 17th Avenue, Mailstop B185, Room 5113C, Aurora, CO, 80045, USA.
| | - Mario A Saporta
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
44
|
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
|
45
|
Martinez NJ, Braisted JC, Dranchak PK, Moran JJ, Larson H, Queme B, Pak E, Dutra A, Rai G, Cheng KCC, Svaren J, Inglese J. Genome-Edited Coincidence and PMP22-HiBiT Fusion Reporter Cell Lines Enable an Artifact-Suppressive Quantitative High-Throughput Screening Strategy for PMP22 Gene-Dosage Disorder Drug Discovery. ACS Pharmacol Transl Sci 2021; 4:1422-1436. [PMID: 34423274 DOI: 10.1021/acsptsci.1c00110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 12/23/2022]
Abstract
Charcot-Marie-Tooth 1A (CMT1A) is the most common form of hereditary peripheral neuropathies, characterized by genetic duplication of the critical myelin gene Peripheral Myelin Protein 22 (PMP22). PMP22 overexpression results in abnormal Schwann cell differentiation, leading to axonal loss and muscle wasting. Since regulation of PMP22 expression is a major target of therapeutic discovery for CMT1A, we sought to establish unbiased approaches that allow the identification of therapeutic agents for this disease. Using genome editing, we generated a coincidence reporter assay that accurately monitors Pmp22 transcript levels in the S16 rat Schwann cell line, while reducing reporter-based false positives. A quantitative high-throughput screen (qHTS) of 42 577 compounds using this assay revealed diverse novel chemical classes that reduce endogenous Pmp22 transcript levels. Moreover, some of these classes show pharmacological specificity in reducing Pmp22 over another major myelin-associated gene, Mpz (Myelin protein zero). Finally, to investigate whether compound-mediated reduction of Pmp22 transcripts translates to reduced PMP22 protein levels, we edited the S16 genome to generate a reporter assay that expresses a PMP22-HiBiT fusion protein using CRISPR/Cas9. Overall, we present a screening platform that combines genome edited cell lines encoding reporters that monitor transcriptional and post-translational regulation of PMP22 with titration-based screening (e.g., qHTS), which could be efficiently incorporated into drug discovery campaigns for CMT1A.
Collapse
Affiliation(s)
- Natalia J Martinez
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - John C Braisted
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Patricia K Dranchak
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - John J Moran
- Department of Comparative Biosciences, and Waisman Center, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Hunter Larson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Bryan Queme
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Evgenia Pak
- National Human Genome Research Institute, National Institute of Health, Bethesda, Maryland 20817, United States
| | - Amalia Dutra
- National Human Genome Research Institute, National Institute of Health, Bethesda, Maryland 20817, United States
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Ken Chih-Chien Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - John Svaren
- Department of Comparative Biosciences, and Waisman Center, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - James Inglese
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States.,National Human Genome Research Institute, National Institute of Health, Bethesda, Maryland 20817, United States
| |
Collapse
|
46
|
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: 1.3] [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
|
47
|
Pisciotta C, Saveri P, Pareyson D. Updated review of therapeutic strategies for Charcot-Marie-Tooth disease and related neuropathies. Expert Rev Neurother 2021; 21:701-713. [PMID: 34033725 DOI: 10.1080/14737175.2021.1935242] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Charcot-Marie-Tooth disease (CMT) and related neuropathies represent the most prevalent inherited neuromuscular disorders. Nonetheless, there is still no pharmacological treatment available for any CMT type. However, the landscape is rapidly evolving and several novel approaches are providing encouraging results in preclinical studies and leading to clinical trials.Areas covered: The authors review the most promising therapies under study and the ongoing/planned clinical trials. Several approaches to address PMP22 overexpression underlying CMT1A, the most frequent subtype, are being tested. Gene silencing, targeting PMP22, and gene therapy, to introduce specific genes or to substitute or modulate defective ones, are being experimented in animal models. Compounds acting on ER stress, unfolded protein response, neuregulin pathways, phosphoinositides metabolism, axonal transport and degeneration, inflammation, polyol pathway, deoxysphingolipid metabolism, purine nucleotide pool are potential therapeutic candidates for different forms of CMT and related neuropathies.Expert opinion: We are getting closer to find effective therapies for CMT, but are far behind the exciting examples of other genetic neuromuscular disorders. The authors analyze the possible reasons for this gap and the way to fill it. Preclinical and clinical research is ongoing with coordinated efforts and they are confident that in the next few years we will see the first effective treatments.
Collapse
Affiliation(s)
- Chiara Pisciotta
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Saveri
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
48
|
Stavrou M, Sargiannidou I, Georgiou E, Kagiava A, Kleopa KA. Emerging Therapies for Charcot-Marie-Tooth Inherited Neuropathies. Int J Mol Sci 2021; 22:6048. [PMID: 34205075 PMCID: PMC8199910 DOI: 10.3390/ijms22116048] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited neuropathies known as Charcot-Marie-Tooth (CMT) disease are genetically heterogeneous disorders affecting the peripheral nerves, causing significant and slowly progressive disability over the lifespan. The discovery of their diverse molecular genetic mechanisms over the past three decades has provided the basis for developing a wide range of therapeutics, leading to an exciting era of finding treatments for this, until now, incurable group of diseases. Many treatment approaches, including gene silencing and gene replacement therapies, as well as small molecule treatments are currently in preclinical testing while several have also reached clinical trial stage. Some of the treatment approaches are disease-specific targeted to the unique disease mechanism of each CMT form, while other therapeutics target common pathways shared by several or all CMT types. As promising treatments reach the stage of clinical translation, optimal outcome measures, novel biomarkers and appropriate trial designs are crucial in order to facilitate successful testing and validation of novel treatments for CMT patients.
Collapse
Affiliation(s)
- Marina Stavrou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Elena Georgiou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Alexia Kagiava
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
| | - Kleopas A. Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.S.); (I.S.); (E.G.); (A.K.)
- Center for Neuromuscular Diseases, The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
| |
Collapse
|
49
|
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: 4.3] [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
|
50
|
Abstract
PURPOSE OF REVIEW Hereditary motor neuropathies (HMN) comprise a broad genotypic and phenotypic spectrum of rare, progressively disabling diseases manifesting with length-dependent muscle weakness and atrophy. To date, more than half of the cases cannot be genetically explained. To provide symptomatic and disease-modifying treatments in the future, a better understanding of disease mechanisms is required. RECENT FINDINGS By whole exome and genome sequencing, the discovery of several novel genes (SCO2, TDRKH, SPTAN1, CADM3, and SORD) involved in the pathogenesis of HMN has now relevantly changed the pathophysiological knowledge. This recent success in causative understanding has mainly been driven by the development of functional models including cell culture, animal, and patient-derived induced pluripotent stem cell platforms. These models have an important impact on therapeutic advances including broader approaches to prevent or reverse axonal degeneration and individualized gene silencing attempts using sequence-specific RNA degradation mechanisms. SUMMARY In rare diseases such as HMN, the recent development of genetic sequencing and data interpretation methods has enabled a broader diagnostic approach, whereas treatment strategies are becoming more individualized. Significant milestones have been reached in the discovery of new genes, the establishment of functional disease models, and the preclinical development of mechanistic-based therapies.
Collapse
|