https://reporter.nih.gov/project-details/10226201
Genomic Studies in Charcot-Marie-Tooth Disease
https://reporter.nih.gov/project-details/10226201
Genomic Studies in Charcot-Marie-Tooth Disease
Creation of a Schwann Cell Gene Regulatory Network
https://reporter.nih.gov/project-details/10466574
https://www.nature.com/articles/s10038-022-01031-2
In this article, we review the epidemiology, genetic diagnosis, and clinicogenetic characteristics of CMT in Japan. In addition, we discuss the newly identified novel causative genes for CMT/IPNs in Japan, namely MME and COA7. Identification of the new causes of CMT will facilitate in-depth characterization of the underlying molecular mechanisms of CMT, leading to the establishment of therapeutic approaches such as drug development and gene therapy.
https://clinicaltrials.gov/ct2/show/NCT03520751
Phase I/IIa Trial Evaluating scAAV1.tMCK.NTF3 for Treatment of Charcot-Marie-Tooth Neuropathy Type 1A (CMT1A)
Researchers at Cyprus Institute of Neurology & Genetics Complete Third Milestone of CMT1A Gene Therapy Project
https://www.ncbi.nlm.nih.gov/pubmed/33419039?dopt=Abstract
https://www.mdpi.com/1422-0067/22/2/491/htm
Epigenetic Regulation of ALS and CMT: A Lesson from Drosophila Models.
Related Articles
Epigenetic Regulation of ALS and CMT: A Lesson from Drosophila Models.
Int J Mol Sci. 2021 Jan 06;22(2):
Authors: Yamaguchi M, Omori K, Asada S, Yoshida H
Abstract
Amyotrophic lateral sclerosis (ALS) is the third most common neurodegenerative disorder and is sometimes associated with frontotemporal dementia. Charcot-Marie-Tooth disease (CMT) is one of the most commonly inherited peripheral neuropathies causing the slow progression of sensory and distal muscle defects. Of note, the severity and progression of CMT symptoms markedly vary. The phenotypic heterogeneity of ALS and CMT suggests the existence of modifiers that determine disease characteristics. Epigenetic regulation of biological functions via gene expression without alterations in the DNA sequence may be an important factor. The methylation of DNA, noncoding RNA, and post-translational modification of histones are the major epigenetic mechanisms. Currently, Drosophila is emerging as a useful ALS and CMT model. In this review, we summarize recent studies linking ALS and CMT to epigenetic regulation with a strong emphasis on approaches using Drosophila models.
PMID: 33419039 [PubMed – as supplied by publisher]
PubMed:33419039
Yamaguchi M, Omori K, Asada S, Yoshida H
https://www.fiercebiotech.com/biotech/neurogene-has-raised-115-million-a-second-round-financing-will-accelerate-its-plans-to
Neurogene adds another $115M to the pot for trials for its gene therapies, including its Charcot-Marie-Tooth disease (CMT) type 4J program
https://academic.oup.com/brain/advance-article-abstract/doi/10.1093/brain/awaa311/5989972?redirectedFrom=fulltext
https://www.ncbi.nlm.nih.gov/pubmed/33210134?dopt=Abstract
The expanding genetic landscape of hereditary motor neuropathies.
Related Articles
The expanding genetic landscape of hereditary motor neuropathies.
Brain. 2020 Nov 19;:
Authors: Beijer D, Baets J
Abstract
Hereditary motor neuropathies are clinically and genetically diverse disorders characterized by length-dependent axonal degeneration of lower motor neurons. Although currently as many as 26 causal genes are known, there is considerable missing heritability compared to other inherited neuropathies such as Charcot-Marie-Tooth disease. Intriguingly, this genetic landscape spans a discrete number of key biological processes within the peripheral nerve. Also, in terms of underlying pathophysiology, hereditary motor neuropathies show striking overlap with several other neuromuscular and neurological disorders. In this review, we provide a current overview of the genetic spectrum of hereditary motor neuropathies highlighting recent reports of novel genes and mutations or recent discoveries in the underlying disease mechanisms. In addition, we link hereditary motor neuropathies with various related disorders by addressing the main affected pathways of disease divided into five major processes: axonal transport, tRNA aminoacylation, RNA metabolism and DNA integrity, ion channels and transporters and endoplasmic reticulum.
PMID: 33210134 [PubMed – as supplied by publisher]
PubMed:33210134
Beijer D, Baets J
https://www.hindawi.com/journals/bmri/2020/1353516/
https://www.ncbi.nlm.nih.gov/pubmed/33029488?dopt=Abstract
Identification of Candidate Genes Associated with Charcot-Marie-Tooth Disease by Network and Pathway Analysis.
Identification of Candidate Genes Associated with Charcot-Marie-Tooth Disease by Network and Pathway Analysis.
Biomed Res Int. 2020;2020:1353516
Authors: Zhong M, Luo Q, Ye T, Zhu X, Chen X, Liu J
Abstract
Charcot-Marie-Tooth Disease (CMT) is the most common clinical genetic disease of the peripheral nervous system. Although many studies have focused on elucidating the pathogenesis of CMT, few focuses on achieving a systematic analysis of biology to decode the underlying pathological molecular mechanisms and the mechanism of its disease remains to be elucidated. So our study may provide further useful insights into the molecular mechanisms of CMT based on a systematic bioinformatics analysis. In the current study, by reviewing the literatures deposited in PUBMED, we identified 100 genes genetically related to CMT. Then, the functional features of the CMT-related genes were examined by R software and KOBAS, and the selected biological process crosstalk was visualized with the software Cytoscape. Moreover, CMT specific molecular network analysis was conducted by the Molecular Complex Detection (MCODE) Algorithm. The biological function enrichment analysis suggested that myelin sheath, axon, peripheral nervous system, mitochondrial function, various metabolic processes, and autophagy played important roles in CMT development. Aminoacyl-tRNA biosynthesis, metabolic pathways, and vasopressin-regulated water reabsorption were significantly enriched in the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway network, suggesting that these pathways may play key roles in CMT occurrence and development. According to the crosstalk, the biological processes could be roughly divided into a correlative module and two separate modules. MCODE clusters showed that in top 3 clusters, 13 of CMT-related genes were included in the network and 30 candidate genes were discovered which might be potentially related to CMT. The study may help to update the new understanding of the pathogenesis of CMT and expand the potential genes of CMT for further exploration.
PMID: 33029488 [PubMed – in process]
PubMed:33029488
Zhong M, Luo Q, Ye T, Zhu X, Chen X, Liu J
https://www.prnewswire.com/news-releases/dtx-pharma-receives-new-funding-grant-to-advance-rna-based-therapeutic-programs-for-charcot-marie-tooth-cmt-301135672.html
DTx Pharma Receives New Funding Grant to Advance RNA-based Therapeutic Programs for Charcot-Marie Tooth (CMT)
-CMT is a genetic rare disease driven by the duplication of the PMP22 gene-
https://link.springer.com/article/10.1007/s00415-020-10171-4
https://www.ncbi.nlm.nih.gov/pubmed/32897397?dopt=Abstract
Deep geno- and phenotyping in two consanguineous families with CMT2 reveals HADHA as an unusual disease-causing gene and an intronic variant in GDAP1 as an unusual mutation.
Related Articles
Deep geno- and phenotyping in two consanguineous families with CMT2 reveals HADHA as an unusual disease-causing gene and an intronic variant in GDAP1 as an unusual mutation.
J Neurol. 2020 Sep 08;:
Authors: Khani M, Taheri H, Shamshiri H, Moazzeni H, Hardy J, Bras JT, InanlooRahatloo K, Alavi A, Nafissi S, Elahi E
Abstract
BACKGROUND: Charcot-Marie-Tooth (CMT) disease is a prevalent and heterogeneous peripheral neuropathy. Most patients affected with the axonal form of CMT (CMT2) do not harbor mutations in the approximately 90 known CMT-associated genes. We aimed to identify causative genes in two CMT2 pedigrees.
METHODS: Neurologic examination, laboratory tests and brain MRIs were performed. Genetic analysis included exome sequencing of four patients from the two pedigrees. The predicted effect of a deep intronic mutation on splicing was tested by regular and real-time PCR and sequencing.
RESULTS: Clinical data were consistent with CMT2 diagnosis. Inheritance patterns were autosomal recessive. Exome data of CMT2-101 did not include mutations in known CMT-associated genes. Sequence data, segregation analysis, bioinformatics analysis, evolutionary conservation, and information in the literature strongly implicated HADHA as the causative gene. An intronic variation positioned 23 nucleotides away from following intron/exon border in GDAP1 was ultimately identified as cause of CMT in CMT2-102. It was shown to affect splicing.
CONCLUSION: The finding of a HADHA mutation as a cause of CMT is of interest because its encoded protein is a subunit of the mitochondrial trifunctional protein (MTP) complex, a mitochondrial enzyme involved in long chain fatty acid oxidation. Long chain fatty acid oxidation is an important source of energy for skeletal muscles. The mutation found in CMT2-102 is only the second intronic mutation reported in GDAP1. The mutation in the CMT2-102 pedigree was outside the canonical splice site sequences, emphasizing the importance of careful examination of available intronic sequences in exome sequence data.
PMID: 32897397 [PubMed – as supplied by publisher]
PubMed:32897397
Khani M, Taheri H, Shamshiri H, Moazzeni H, Hardy J, Bras JT, InanlooRahatloo K, Alavi A, Nafissi S, Elahi E
https://pubmed.ncbi.nlm.nih.gov/32815244/
https://www.ncbi.nlm.nih.gov/pubmed/32815244?dopt=Abstract
Expanding the phenotypic spectrum of TRIM2-associated Charcot-Marie-Tooth disease.
Related Articles
Expanding the phenotypic spectrum of TRIM2-associated Charcot-Marie-Tooth disease.
J Peripher Nerv Syst. 2020 Aug 19;:
Authors: Magri S, Danti FR, Balistreri F, Baratta S, Ciano C, Pagliano E, Taroni F, Moroni I
Abstract
BACKGROUND AND AIMS: Charcot-Marie-Tooth disease (CMT) is a clinically and genetically heterogeneous group of distal symmetric polyneuropathies due to progressive and length-dependent degeneration of peripheral nerves. Cranial nerve involvement has been described in association with various CMT-genes mutations, such as GDAP1, TRPV4, MFN2, MTMR2 and EGR2. Compound heterozygous mutations in the TRIM2 gene, encoding an E3 ubiquitin ligase, were previously identified in two patients with early-onset axonal CMT (CMT2). One of them also had bilateral vocal cord paralysis. The aim of this study is to further delineate the phenotypic and molecular genetic features of TRIM2-related CMT.
METHODS: We studied clinical, genetic and neurophysiological aspects of two unrelated CMT2 patients. Genetic analysis was performed by next generation sequencing of a multigene CMT panel.
RESULTS: Patients presented with congenital hypotonia and bilateral clubfoot, delayed motor milestones, and severely progressive axonal neuropathy. Interestingly, along with vocal cord paralysis, they exhibited clinical features secondary to the involvement of several other cranial nerves, such as facial weakness, dysphagia, dyspnoea and acoustic impairment. Genetic analysis revealed two novel TRIM2 mutations in each patient.
INTERPRETATION: Our results expand the genotypic and phenotypic spectrum of TRIM2 deficiency showing that cranial nerves involvement is a core feature in this CMT2-subtype. Its finding should prompt physicians to suspect TRIM2 neuropathy. Conversely, patients carrying TRIM2 variants should be carefully evaluated for the presence of cranial nerve dysfunction in order to prevent and manage its impact on auditory and respiratory function and nutrition. This article is protected by copyright. All rights reserved.
PMID: 32815244 [PubMed – as supplied by publisher]
PubMed:32815244
Magri S, Danti FR, Balistreri F, Baratta S, Ciano C, Pagliano E, Taroni F, Moroni I
https://www.nature.com/articles/s41419-020-02798-y
https://pubmed.ncbi.nlm.nih.gov/32703932/
Developmental demands contribute to early neuromuscular degeneration in CMT2D mice
James N. Sleigh, Aleksandra M. Mech & Giampietro Schiavo
Cell Death & Disease volume 11, Article number: 564 (2020) Cite this article
78 Accesses
9 Altmetric
Metricsdetails
Abstract
Dominantly inherited, missense mutations in the widely expressed housekeeping gene, GARS1, cause Charcot-Marie-Tooth type 2D (CMT2D), a peripheral neuropathy characterised by muscle weakness and wasting in limb extremities. Mice modelling CMT2D display early and selective neuromuscular junction (NMJ) pathology, epitomised by disturbed maturation and neurotransmission, leading to denervation. Indeed, the NMJ disruption has been reported in several different muscles; however, a systematic comparison of neuromuscular synapses from distinct body locations has yet to be performed. We therefore analysed NMJ development and degeneration across five different wholemount muscles to identify key synaptic features contributing to the distinct pattern of neurodegeneration in CMT2D mice. Denervation was found to occur along a distal-to-proximal gradient, providing a cellular explanation for the greater weakness observed in mutant Gars hindlimbs compared with forelimbs. Nonetheless, muscles from similar locations and innervated by axons of equivalent length showed significant differences in neuropathology, suggestive of additional factors impacting on site-specific neuromuscular degeneration. Defective NMJ development preceded and associated with degeneration, but was not linked to a delay of wild-type NMJ maturation processes. Correlation analyses indicate that muscle fibre type nor synaptic architecture explain the differential denervation of CMT2D NMJs, rather it is the extent of post-natal synaptic growth that predisposes to neurodegeneration. Together, this work improves our understanding of the mechanisms driving synaptic vulnerability in CMT2D and hints at pertinent pathogenic pathways.
https://www.translationalres.com/article/S1931-5244(20)30175-4/pdf
https://www.ncbi.nlm.nih.gov/pubmed/32693030?dopt=Abstract
Treating PMP22 gene duplication-related Charcot-Marie-Tooth disease: The past, the present and the future.
Treating PMP22 gene duplication-related Charcot-Marie-Tooth disease: The past, the present and the future.
Transl Res. 2020 Jul 18;:
Authors: Boutary S, Echaniz-Laguna A, Adams D, Loisel-Duwattez J, Schumacher M, Massaad C, Massaad-Massade L
Abstract
Charcot-Marie-Tooth (CMT) disease is the most frequent inherited neuropathy, affecting 1/1500 to 1/10000. CMT1A represents 60-70% of all CMT and is caused by a duplication on chromosome 17p11.2 leading to an overexpression of the Peripheral Myelin Protein 22 (PMP22). PMP22 gene is under tight regulation and small changes in its expression influences myelination and affect motor and sensory functions. To date, CMT1A treatment is symptomatic and classic pharmacological options have been disappointing. Here, we review the past, present and future treatment options for CMT1A, with a special emphasis on the highly promising potential of PMP22-targeted small interfering RNA (siRNA) and antisense oligonucleotides (ASO).
PMID: 32693030 [PubMed – as supplied by publisher]
PubMed:32693030
Boutary S, Echaniz-Laguna A, Adams D, Loisel-Duwattez J, Schumacher M, Massaad C, Massaad-Massade L
https://onlinelibrary.wiley.com/doi/abs/10.1111/jns.12395
https://www.ncbi.nlm.nih.gov/pubmed/32639100?dopt=Abstract
A novel HSPB1 mutation associated with a late onset CMT2 phenotype: Case presentation and systematic review of the literature.
A novel HSPB1 mutation associated with a late onset CMT2 phenotype: Case presentation and systematic review of the literature.
J Peripher Nerv Syst. 2020 Jul 08;:
Authors: Taga A, Cornblath DR
Abstract
Mutations in the HSPB1 gene are associated with Charcot-Marie-Tooth (CMT) disease type 2F (CMT2F) and distal hereditary motor neuropathy type 2 (dHMN2). More than 18 pathogenic mutations spanning across the whole HSPB1 gene have been reported. Three family members with a novel p.P57S (c.169C>T) HSPB1 mutation resulting in a late onset axonal neuropathy with heterogeneous clinical and electrophysiological features are detailed. We systematically reviewed published case reports and case series on HSPB1 mutations. While a genotype-phenotype correlation was not obvious, we identified a common phenotype, which included adult onset, male predominance, motor more frequently than sensory involvement, distal and symmetric distribution with preferential involvement of plantar flexors, and a motor and axonal electrophysiological picture.
PMID: 32639100 [PubMed – as supplied by publisher]
PubMed:32639100
Taga A, Cornblath DR
https://www.nature.com/articles/s41598-020-66266-5
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7275085/
https://www.ncbi.nlm.nih.gov/pubmed/32504000?dopt=Abstract
Energy metabolism and mitochondrial defects in X-linked Charcot-Marie-Tooth (CMTX6) iPSC-derived motor neurons with the p.R158H PDK3 mutation.
Related Articles
Energy metabolism and mitochondrial defects in X-linked Charcot-Marie-Tooth (CMTX6) iPSC-derived motor neurons with the p.R158H PDK3 mutation.
Sci Rep. 2020 Jun 05;10(1):9262
Authors: Perez-Siles G, Cutrupi A, Ellis M, Screnci R, Mao D, Uesugi M, Yiu EM, Ryan MM, Choi BO, Nicholson G, Kennerson ML
Abstract
Charcot-Marie-Tooth (CMT) is a group of inherited diseases clinically and genetically heterogenous, characterised by length dependent degeneration of axons of the peripheral nervous system. A missense mutation (p.R158H) in the pyruvate dehydrogenase kinase 3 gene (PDK3) has been identified as the genetic cause for an X-linked form of CMT (CMTX6) in two unrelated families. PDK3 is one of four PDK isoenzymes that regulate the activity of the pyruvate dehydrogenase complex (PDC). The balance between kinases (PDKs) and phosphatases (PDPs) determines the extend of oxidative decarboxylation of pyruvate to generate acetyl CoA, critically linking glycolysis and the energy producing Krebs cycle. We had shown the p.R158H mutation causes hyperactivity of PDK3 and CMTX6 fibroblasts show hyperphosphorylation of PDC, leading to reduced PDC activity and ATP production. In this manuscript we have generated induced pluripotent stem cells (iPSCs) by re-programming CMTX6 fibroblasts (iPSCCMTX6). We also have engineered an isogenic control (iPSCisogenic) and demonstrated that genetic correction of the p.R158H mutation reverses the CMTX6 phenotype. Patient-derived motor neurons (MNCMTX6) show increased phosphorylation of the PDC, energy metabolism defects and mitochondrial abnormalities, including reduced velocity of trafficking mitochondria in the affected axons. Treatment of the MNCMTX6 with a PDK inhibitor reverses PDC hyperphosphorylation and the associated functional deficits founds in the patient motor neurons, demonstrating that the MNCMTX6 and MNisogenic motor neurons provide an excellent neuronal system for compound screening approaches to identify drugs for the treatment of CMTX6.
PMID: 32504000 [PubMed – as supplied by publisher]
PubMed:32504000
Perez-Siles G, Cutrupi A, Ellis M, Screnci R, Mao D, Uesugi M, Yiu EM, Ryan MM, Choi BO, Nicholson G, Kennerson ML
https://www.globenewswire.com/news-release/2020/05/11/2030949/0/en/Sarepta-Therapeutics-and-Dyno-Therapeutics-Announce-Agreement-to-Develop-Next-Generation-Gene-Therapy-Vectors-for-Muscle-Diseases.html
Sarepta Therapeutics and Dyno Therapeutics Announce Agreement to Develop Next-Generation Gene Therapy Vectors for Muscle Diseases
https://onlinelibrary.wiley.com/doi/abs/10.1111/jns.12377
https://www.ncbi.nlm.nih.gov/pubmed/32319184?dopt=Abstract
Genetic spectrum of MCM3AP and its relationship with phenotype of Charcot-Marie-Tooth disease.
Genetic spectrum of MCM3AP and its relationship with phenotype of Charcot-Marie-Tooth disease.
J Peripher Nerv Syst. 2020 Apr 21;:
Authors: Dong HL, Wei Q, Li JQ, Li HF, Bai G, Ma H, Wu ZY
Abstract
BACKGROUND AND AIMS: Mutations in MCM3AP have recently been reported to cause autosomal recessive Charcot-Marie-Tooth disease (CMT). However, only 9 CMT families with MCM3AP mutations have been reported and genotype-phenotype correlation remains unclear. This study aimed to investigate the genetic spectrum of MCM3AP and its relationship with phenotype of CMT.
METHODS: Whole exome sequencing (WES) was performed in the family and variants were validated by Sanger sequencing. Reverse transcription-PCR (RT-PCR) were performed in splicing analysis.
RESULTS: We reported a novel splicing variant (c.5634-1G > T) and a known missense variant (c.2633G > A, p.Arg878His). Functional studies showed that c.5634-1G > T led to splicing defect and aberrant transcript eliminated by nonsense-mediated mRNA decay. The symptom of the patient was less severe and slowly progressed with axonal peripheral neuropathy compared to the reported CMT patients. Genotype-phenotype correlation analysis indicated that affected individuals with null mutations presented with delayed independent walking. The percentage of intellectual disability and loss of ambulation in the null group tended to be greater, although this failed to reach statistical significance.
INTERPRETATION: Our findings expand the genetic spectrum of MCM3AP and suggest that genotype-phenotype correlation would help genetic counseling of MCM3AP in CMT patients. This article is protected by copyright. All rights reserved.
PMID: 32319184 [PubMed – as supplied by publisher]
PubMed:32319184
Dong HL, Wei Q, Li JQ, Li HF, Bai G, Ma H, Wu ZY
https://www.mdpi.com/2073-4409/9/4/1028
https://www.ncbi.nlm.nih.gov/pubmed/32326241?dopt=Abstract
Charcot-Marie-Tooth Type 2B: A New Phenotype Associated with a Novel RAB7A Mutation and Inhibited EGFR Degradation.
Charcot-Marie-Tooth Type 2B: A New Phenotype Associated with a Novel RAB7A Mutation and Inhibited EGFR Degradation.
Cells. 2020 Apr 21;9(4):
Authors: Saveri P, De Luca M, Nisi V, Pisciotta C, Romano R, Piscosquito G, Reilly MM, Polke JM, Cavallaro T, Fabrizi GM, Fossa P, Cichero E, Lombardi R, Lauria G, Magri S, Taroni F, Pareyson D, Bucci C
Abstract
The rare autosomal dominant Charcot-Marie-Tooth type 2B (CMT2B) is associated with mutations in the RAB7A gene, involved in the late endocytic pathway. CMT2B is characterized by predominant sensory loss, ulceromutilating features, with lesser-to-absent motor deficits. We characterized clinically and genetically a family harboring a novel pathogenic RAB7A variant and performed structural and functional analysis of the mutant protein. A 39-year-old woman presented with early-onset walking difficulties, progressive distal muscle wasting and weakness in lower limbs and only mild sensory signs. Electrophysiology demonstrated an axonal sensorimotor neuropathy. Nerve biopsy showed a chronic axonal neuropathy with moderate loss of all caliber myelinated fibers. Next-generation sequencing (NGS) technology revealed in the proband and in her similarly affected father the novel c.377A>G (p.K126R) heterozygous variant predicted to be deleterious. The mutation affects the biochemical properties of RAB7 GTPase, causes altered interaction with peripherin, and inhibition of neurite outgrowth, as for previously reported CMT2B mutants. However, it also shows differences, particularly in the epidermal growth factor receptor degradation process. Altogether, our findings indicate that this RAB7A variant is pathogenic and widens the phenotypic spectrum of CMT2B to include predominantly motor CMT2. Alteration of the receptor degradation process might explain the different clinical presentations in this family.
PMID: 32326241 [PubMed – in process]
PubMed:32326241
Saveri P, De Luca M, Nisi V, Pisciotta C, Romano R, Piscosquito G, Reilly MM, Polke JM, Cavallaro T, Fabrizi GM, Fossa P, Cichero E, Lombardi R, Lauria G, Magri S, Taroni F, Pareyson D, Bucci C
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7164973/
https://www.ncbi.nlm.nih.gov/pubmed/32337334?dopt=Abstract
Novel EGR2 variant that associates with Charcot-Marie-Tooth disease when combined with lipopolysaccharide-induced TNF-α factor T49M polymorphism.
Related Articles
Novel EGR2 variant that associates with Charcot-Marie-Tooth disease when combined with lipopolysaccharide-induced TNF-α factor T49M polymorphism.
Neurol Genet. 2020 Apr;6(2):e407
Authors: Blanco-Cantó ME, Patel N, Velasco-Aviles S, Casillas-Bajo A, Salas-Felipe J, García-Escrivá A, Díaz-Marín C, Cabedo H
Abstract
Objective: To identify novel genetic mechanisms causing Charcot-Marie-Tooth (CMT) disease.
Methods: We performed a next-generation sequencing study of 34 genes associated with CMT in a patient with peripheral neuropathy.
Results: We found a non-previously described mutation in EGR2 (p.P397H). P397H mutation is located within the loop that connects zinc fingers 2 and 3, a pivotal domain for the activity of this transcription factor. Using promoter activity luciferase assays, we found that this mutation promotes decreased transcriptional activity of EGR2. In this patient, we also found a previously described nonpathogenic polymorphism in lipopolysaccharide-induced TNF-α factor (LITAF) (p.T49M). We show that the p.T49M mutation decreases the steady-state levels of the LITAF protein in Schwann cells. Loss of function of LITAF has been shown to produce deregulation in the NRG1-erbB signaling, a pivotal pathway for EGR2 expression by Schwann cells. Surprisingly, our segregation study demonstrates that p.P397H mutation in EGR2 is not sufficient to produce CMT disease. Most notably, only those patients expressing simultaneously the LITAF T49M polymorphism develop peripheral neuropathy.
Conclusions: Our data support that the LITAF loss-of-function interferes with the expression of the transcriptional-deficient EGR2 P397H mutant hampering Schwann cell differentiation and suggest that in vivo both genes act in tandem to allow the proper development of myelin.
PMID: 32337334 [PubMed]
PubMed:32337334
Blanco-Cantó ME, Patel N, Velasco-Aviles S, Casillas-Bajo A, Salas-Felipe J, García-Escrivá A, Díaz-Marín C, Cabedo H
https://cmtrf.org/blog/cmt-research-foundation-funds-shift-pharmaceuticals-to-advance-search-for-cmt1a-treatment/
CMT Research Foundation Funds Shift Pharmaceuticals to Advance Search for CMT1A Treatment
Shift to Develop and Analyze Novel Series of Drugs Designed to Control Expression of PMP22 Gene
https://www.biorxiv.org/content/10.1101/2020.01.29.924605v1.full
Long term AAV2/9-mediated silencing of PMP22 prevents CMT1A disease in rats and validates skin biomarkers as treatment outcome measure
Benoit Gautier, Helene Hajjar, Sylvia Soares, Jade Berthelot, Marie Deck, Scarlette Abbou, Graham Campbell, Claire-Maelle Fovet, Vlad Schütza, Antoine Jouvenel, Cyril Rivat, Michel Zerah, Virginie François Le Ravazet, Caroline Le Guiner, Patrick Aubourg, View ORCID ProfileRobert Fledrich, View ORCID ProfileNicolas Tricaud
doi: https://doi.org/10.1101/2020.01.29.924605
https://www.sciencedirect.com/science/article/abs/pii/S0006899320300391?via%3Dihub
https://www.ncbi.nlm.nih.gov/pubmed/32001243?dopt=Abstract
Gene Therapies for Axonal Neuropathies: Available Strategies, Successes to Date, and What to Target Next.
Related Articles
Gene Therapies for Axonal Neuropathies: Available Strategies, Successes to Date, and What to Target Next.
Brain Res. 2020 Jan 27;:146683
Authors: Morelli KH, Hatton CL, Harper SQ, Burgess RW
Abstract
Nearly one-hundred loci in the human genome have been associated with different forms of Charcot-Marie-Tooth disease (CMT) and related inherited neuropathies. Despite this wealth of gene targets, treatment options are still extremely limited, and clear “druggable” pathways are not obvious for many of these mutations. However, recent advances in gene therapies are beginning to circumvent this challenge. Each type of CMT is a monogenic disorder, and the cellular targets are usually well-defined and typically include peripheral neurons or Schwann cells. In addition, the genetic mechanism is often also clear, with loss-of-function mutations requiring restoration of gene expression, and gain-of-function or dominant-negative mutations requiring silencing of the mutant allele. These factors combine to make CMT a good target for developing genetic therapies. Here we will review the state of relatively established gene therapy approaches, including viral vector-mediated gene replacement and antisense oligonucleotides for exon skipping, altering splicing, and gene knockdown. We will also describe earlier stage approaches for allele-specific knockdown and CRIPSR/Cas9 gene editing. We will next describe how these various approaches have been deployed in clinical and preclinical studies. Finally, we will evaluate various forms of CMT as candidates for gene therapy based on the current understanding of their genetics, cellular/tissue targets, validated animal models, and availability of patient populations and natural history data.
PMID: 32001243 [PubMed – as supplied by publisher]
PubMed:32001243
Morelli KH, Hatton CL, Harper SQ, Burgess RW
https://cmtrf.org/blog/partnership-with-dtx-pharma-to-advance-gene-therapy-development-for-cmt1a/
A new partnership to optimize antisense oligonucleotides as a gene therapy for CMT1A
CMTRF partners with DTx Pharma to optimize antisense oligonucleotides as a gene therapy for CMT1A
Dec 20, 2019 | CMT Research News, CMTRF Funded Research, Gene Therapy, Press Releases, Research news, Therapy Types | 0 comments
ATLANTA (December 20, 2019) The CMT Research Foundation (CMTRF), a nonprofit focused solely on delivering treatments and cures for Charcot-Marie-Tooth, today announced it has partnered with DTx Pharma, an RNA medicines company breaking open new therapeutic areas for oligonucleotide (short DNA or RNA molecules) drugs by overcoming the delivery challenge that has limited the utility of this promising new potential gene therapy for CMT1A.
https://www.nature.com/articles/s10038-019-0710-5
https://www.ncbi.nlm.nih.gov/pubmed/31852984?dopt=Abstract
Identification of novel pathogenic copy number variations in Charcot-Marie-Tooth disease.
Related Articles
Identification of novel pathogenic copy number variations in Charcot-Marie-Tooth disease.
J Hum Genet. 2019 Dec 18;:
Authors: Mortreux J, Bacquet J, Boyer A, Alazard E, Bellance R, Giguet-Valard AG, Cerino M, Krahn M, Audic F, Chabrol B, Laugel V, Desvignes JP, Béroud C, Nguyen K, Verschueren A, Lévy N, Attarian S, Delague V, Missirian C, Bonello-Palot N
Abstract
Charcot-Marie-Tooth disease (CMT) is a hereditary sensory-motor neuropathy characterized by a strong clinical and genetic heterogeneity. Over the past few years, with the occurrence of whole-exome sequencing (WES) or whole-genome sequencing (WGS), the molecular diagnosis rate has been improved by allowing the screening of more than 80 genes at one time. In CMT, except the recurrent PMP22 duplication accounting for about 60% of pathogenic variations, pathogenic copy number variations (CNVs) are rarely reported and only a few studies screening specifically CNVs have been performed. The aim of the present study was to screen for CNVs in the most prevalent genes associated with CMT in a cohort of 200 patients negative for the PMP22 duplication. CNVs were screened using the Exome Depth software on next generation sequencing (NGS) data obtained by targeted capture and sequencing of a panel of 81 CMT associated genes. Deleterious CNVs were identified in four patients (2%), in four genes: GDAP1, LRSAM1, GAN, and FGD4. All CNVs were confirmed by high-resolution oligonucleotide array Comparative Genomic Hybridization (aCGH) and/or quantitative PCR. By identifying four new CNVs in four different genes, we demonstrate that, although they are rare mutational events in CMT, CNVs might contribute significantly to mutational spectrum of Charcot-Marie-Tooth disease and should be searched in routine NGS diagnosis. This strategy increases the molecular diagnosis rate of patients with neuropathy.
PMID: 31852984 [PubMed – as supplied by publisher]
PubMed:31852984
Mortreux J, Bacquet J, Boyer A, Alazard E, Bellance R, Giguet-Valard AG, Cerino M, Krahn M, Audic F, Chabrol B, Laugel V, Desvignes JP, Béroud C, Nguyen K, Verschueren A, Lévy N, Attarian S, Delague V, Missirian C, Bonello-Palot N
https://www.nature.com/articles/s41598-019-55875-4
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6920433/
https://www.ncbi.nlm.nih.gov/pubmed/31852952?dopt=Abstract
EGR2 mutation causes severe early-onset Charcot-Marie-Tooth Neuropathy Type 3 (Dejerine-Sottas Neuropathy).
A de novo EGR2 variant, c.1232A > G p.Asp411Gly, causes severe early-onset Charcot-Marie-Tooth Neuropathy Type 3 (Dejerine-Sottas Neuropathy).
Related Articles
A de novo EGR2 variant, c.1232A > G p.Asp411Gly, causes severe early-onset Charcot-Marie-Tooth Neuropathy Type 3 (Dejerine-Sottas Neuropathy).
Sci Rep. 2019 Dec 18;9(1):19336
Authors: Grosz BR, Golovchenko NB, Ellis M, Kumar K, Nicholson GA, Antonellis A, Kennerson ML
Abstract
EGR2 (early growth response 2) is a crucial transcription factor for the myelination of the peripheral nervous system. Mutations in EGR2 are reported to cause a heterogenous spectrum of peripheral neuropathy with wide variation in both severity and age of onset, including demyelinating and axonal forms of Charcot-Marie Tooth (CMT) neuropathy, Dejerine-Sottas neuropathy (DSN/CMT3), and congenital hypomyelinating neuropathy (CHN/CMT4E). Here we report a sporadic de novo EGR2 variant, c.1232A > G (NM_000399.5), causing a missense p.Asp411Gly substitution and discovered through whole-exome sequencing (WES) of the proband. The resultant phenotype is severe demyelinating DSN with onset at two years of age, confirmed through nerve biopsy and electrophysiological examination. In silico analyses showed that the Asp411 residue is evolutionarily conserved, and the p.Asp411Gly variant was predicted to be deleterious by multiple in silico analyses. A luciferase-based reporter assay confirmed the reduced ability of p.Asp411Gly EGR2 to activate a PMP22 (peripheral myelin protein 22) enhancer element compared to wild-type EGR2. This study adds further support to the heterogeneity of EGR2-related peripheral neuropathies and provides strong functional evidence for the pathogenicity of the p.Asp411Gly EGR2 variant.
PMID: 31852952 [PubMed – in process]
PubMed:31852952
Grosz BR, Golovchenko NB, Ellis M, Kumar K, Nicholson GA, Antonellis A, Kennerson ML
https://www.ncbi.nlm.nih.gov/pubmed/31790684?dopt=Abstract
https://www.sciencedirect.com/science/article/abs/pii/S0006899319306262?via%3Dihub
Gene therapy approaches targeting Schwann cells for demyelinating neuropathies.
Related Articles
Gene therapy approaches targeting Schwann cells for demyelinating neuropathies.
Brain Res. 2019 Nov 29;:146572
Authors: Sargiannidou I, Kagiava A, Kleopa KA
Abstract
Charcot-Marie-Tooth disease (CMT) encompasses numerous genetically heterogeneous inherited neuropathies, which together are one of the commonest neurogenetic disorders. Axonal CMT types result from mutations in neuronally expressed genes, whereas demyelinating CMT forms mostly result from mutations in genes expressed by myelinating Schwann cells. The demyelinating forms are the most common, and may be caused by dominant mutations and gene dosage effects (as in CMT1), as well as by recessive mutations and loss of function mechanisms (as in CMT4). The discovery of causative genes and increasing insights into molecular mechanisms through the study of experimental disease models has provided the basis for the development of gene therapy approaches. For demyelinating CMT, gene silencing or gene replacement strategies need to be targeted to Schwann cells. Progress in gene replacement for two different CMT forms, including CMT1X caused by GJB1 gene mutations, and CMT4C, caused by SH3TC2 gene mutations, has been made through the use of a myelin-specific promoter to restrict expression in Schwann cells, and by lumbar intrathecal delivery of lentiviral viral vectors to achieve more widespread biodistribution in the peripheral nervous system. This review summarizes the molecular-genetic mechanisms of selected demyelinating CMT neuropathies and the progress made so far, as well as the remaining challenges in the path towards a gene therapy to treat these disorders through the use of optimal gene therapy tools such as using clinically translatable delivery methods and adeno-associated viral (AAV) vectors.
PMID: 31790684 [PubMed – as supplied by publisher]
PubMed:31790684
Sargiannidou I, Kagiava A, Kleopa KA
https://www.sciencedaily.com/releases/2019/11/191121121730.htm
New mechanism of neurodegeneration
Date:
November 21, 2019
Source:
VIB (the Flanders Institute for Biotechnology)
Summary:
Charcot-Marie-Tooth disease (CMT) is an inherited neurodegenerative condition that affects 1 in 2500 individuals. Currently, however, it is still lacking effective treatment options. New research has demonstrated that a class of cytoplasmic enzymes called tRNA synthetases can cause CMT by interfering with the gene transcription in the nucleus.
https://academic.oup.com/nar/advance-article/doi/10.1093/nar/gkz1070/5622709
https://www.ncbi.nlm.nih.gov/pubmed/31713617?dopt=Abstract
Targeted PMP22 TATA-box editing by CRISPR/Cas9 reduces demyelinating neuropathy of Charcot-Marie-Tooth disease type 1A in mice.
Targeted PMP22 TATA-box editing by CRISPR/Cas9 reduces demyelinating neuropathy of Charcot-Marie-Tooth disease type 1A in mice.
Nucleic Acids Res. 2019 Nov 12;:
Authors: Lee JS, Lee JY, Song DW, Bae HS, Doo HM, Yu HS, Lee KJ, Kim HK, Hwang H, Kwak G, Kim D, Kim S, Hong YB, Lee JM, Choi BO
Abstract
Charcot-Marie-Tooth 1A (CMT1A) is the most common inherited neuropathy without a known therapy, which is caused by a 1.4 Mb duplication on human chromosome 17, which includes the gene encoding the peripheral myelin protein of 22 kDa (PMP22). Overexpressed PMP22 protein from its gene duplication is thought to cause demyelination and subsequently axonal degeneration in the peripheral nervous system (PNS). Here, we targeted TATA-box of human PMP22 promoter to normalize overexpressed PMP22 level in C22 mice, a mouse model of CMT1A harboring multiple copies of human PMP22. Direct local intraneural delivery of CRISPR/Cas9 designed to target TATA-box of PMP22 before the onset of disease, downregulates gene expression of PMP22 and preserves both myelin and axons. Notably, the same approach was effective in partial rescue of demyelination even after the onset of disease. Collectively, our data present a proof-of-concept that CRISPR/Cas9-mediated targeting of TATA-box can be utilized to treat CMT1A.
PMID: 31713617 [PubMed – as supplied by publisher]
PubMed:31713617
Lee JS, Lee JY, Song DW, Bae HS, Doo HM, Yu HS, Lee KJ, Kim HK, Hwang H, Kwak G, Kim D, Kim S, Hong YB, Lee JM, Choi BO
https://www.sciencedirect.com/science/article/abs/pii/S0006899319305864?via%3Dihub
https://www.ncbi.nlm.nih.gov/pubmed/31678418?dopt=Abstract
Overlapping spectrums: the clinicogenetic commonalities between Charcot-Marie-Tooth and other neurodegenerative diseases.
Related Articles
Overlapping spectrums: the clinicogenetic commonalities between Charcot-Marie-Tooth and other neurodegenerative diseases.
Brain Res. 2019 Oct 31;:146532
Authors: Martin PB, Hicks AN, Holbrook SE, Cox GA
Abstract
Charcot-Marie-Tooth (CMT) disease is a progressive and heterogeneous inherited peripheral neuropathy. A myriad of genetic factors have been identified that contribute to the degeneration of motor and sensory axons in a length-dependent manner. Emerging biological themes underlying disease include defects in axonal trafficking, dysfunction in RNA metabolism and protein homeostasis, as well deficits in the cellular stress response. Moreover, genetic contributions to CMT can have overlap with other neuropathies, motor neuron diseases (MNDs) and neurodegenerative disorders. Recent progress in understanding the molecular biology of CMT and overlapping syndromes aids in the search for necessary therapeutic targets.
PMID: 31678418 [PubMed – as supplied by publisher]
PubMed:31678418
Martin PB, Hicks AN, Holbrook SE, Cox GA
https://www.sciencedirect.com/science/article/abs/pii/S0006899319305876?via%3Dihub
https://www.ncbi.nlm.nih.gov/pubmed/31669284?dopt=Abstract
Gene therapy to promote regeneration in Charcot-Marie-Tooth disease.
Related Articles
Gene therapy to promote regeneration in Charcot-Marie-Tooth disease.
Brain Res. 2019 Oct 24;:146533
Authors: Sahenk Z, Ak BO
Abstract
The molecular pathogenesis underlying Charcot-Marie-Tooth (CMT) neuropathy subtypes is becoming increasingly variable and identification of common approaches for treatment, independently of the disease causing gene defect, is therefore much desirable. Gene therapy approach from the clinical translational view point is particularly challenging for the most common “demyelinating” CMT1 subtypes, caused by primary Schwann cell genetic defects. Studies have shown that impaired regenerative capacity of distal axons is major contributing factor to distal axonal loss in primary Schwann cell genetic defects and neurotrophin 3 (NT-3) improves impaired regeneration in CMT1 mouse models. This review surveys the evidence supporting the rationale for AAV1.NT-3 surrogate gene therapy to improve nerve regeneration in CMT1A. The translational process, from proof of principal studies to the design of the phase I/IIa trial evaluating scAAV1.tMCK.NTF3 gene therapy for treatment of CMT1A is summarized.
PMID: 31669284 [PubMed – as supplied by publisher]
PubMed:31669284
Sahenk Z, Ak BO
https://cmtrf.org/blog/emerging-landscape-of-modifier-genes-in-cmt/
Emerging Landscape of Modifier Genes in CMT
https://www.nature.com/articles/s41582-019-0254-5
https://www.ncbi.nlm.nih.gov/pubmed/31582811?dopt=Abstract
Next-generation sequencing in Charcot-Marie-Tooth disease: opportunities and challenges.
Related Articles
Next-generation sequencing in Charcot-Marie-Tooth disease: opportunities and challenges.
Nat Rev Neurol. 2019 Oct 03;:
Authors: Pipis M, Rossor AM, Laura M, Reilly MM
Abstract
Charcot-Marie-Tooth disease and the related disorders hereditary motor neuropathy and hereditary sensory neuropathy, collectively termed CMT, are the commonest group of inherited neuromuscular diseases, and they exhibit wide phenotypic and genetic heterogeneity. CMT is usually characterized by distal muscle atrophy, often with foot deformity, weakness and sensory loss. In the past decade, next-generation sequencing (NGS) technologies have revolutionized genomic medicine and, as these technologies are being applied to clinical practice, they are changing our diagnostic approach to CMT. In this Review, we discuss the application of NGS technologies, including disease-specific gene panels, whole-exome sequencing, whole-genome sequencing (WGS), mitochondrial sequencing and high-throughput transcriptome sequencing, to the diagnosis of CMT. We discuss the growing challenge of variant interpretation and consider how the clinical phenotype can be combined with genetic, bioinformatic and functional evidence to assess the pathogenicity of genetic variants in patients with CMT. WGS has several advantages over the other techniques that we discuss, which include unparalleled coverage of coding, non-coding and intergenic areas of both nuclear and mitochondrial genomes, the ability to identify structural variants and the opportunity to perform genome-wide dense homozygosity mapping. We propose an algorithm for incorporating WGS into the CMT diagnostic pathway.
PMID: 31582811 [PubMed – as supplied by publisher]
PubMed:31582811
Pipis M, Rossor AM, Laura M, Reilly MM
https://www.sciencedirect.com/science/article/abs/pii/S0006899319305451?via%3Dihub
https://www.ncbi.nlm.nih.gov/pubmed/31586623?dopt=Abstract
Regulating PMP22 Expression as a Dosage Sensitive Neuropathy Gene.
Regulating PMP22 Expression as a Dosage Sensitive Neuropathy Gene.
Brain Res. 2019 Oct 03;:146491
Authors: Pantera H, Shy ME, Svaren J
Abstract
Structural variation in the human genome has emerged as a major cause of disease as genomic data have accumulated. One of the most common structural variants associated with human disease causes the heritable neuropathy known as Charcot-Marie-Tooth (CMT) disease type 1A. This 1.4 Mb duplication causes nearly half of the CMT cases that are genetically diagnosed. The PMP22 gene is highly induced in Schwann cells during development, although its precise role in myelin formation and homeostasis is still under active investigation. The PMP22 gene can be considered as a nucleoprotein complex with enzymatic activity to produce the PMP22 transcript, and the complex is allosterically regulated by transcription factors that respond to intracellular signals and epigenomic modifications. The control of PMP22 transcript levels has been one of the major therapeutic targets of therapy development, and this review summarizes those approaches as well as efforts to characterize the regulation of the PMP22 gene.
PMID: 31586623 [PubMed – as supplied by publisher]
PubMed:31586623
Pantera H, Shy ME, Svaren J
https://www.jci.org/articles/view/130600
https://www.ncbi.nlm.nih.gov/pubmed/31557132?dopt=Abstract
Efficacy of allele-specific RNAi as a potential therapeutic for Charcot-Marie-Tooth type 2D (CMT2D)
Allele-specific RNA interference prevents neuropathy in Charcot-Marie-Tooth disease type 2D mouse models.
Allele-specific RNA interference prevents neuropathy in Charcot-Marie-Tooth disease type 2D mouse models.
J Clin Invest. 2019 Sep 26;:
Authors: Morelli KH, Griffin LB, Pyne NK, Wallace LM, Fowler AM, Oprescu SN, Takase R, Wei N, Meyer-Schuman R, Mellacheruvu D, Kitzman JO, Kocen SG, Hines TJ, Spaulding EL, Lupski JR, Nesvizhskii A, Mancias P, Butler IJ, Yang XL, Hou YM, Antonellis A, Harper SQ, Burgess RW
Abstract
Gene therapy approaches are being deployed to treat recessive genetic disorders by restoring the expression of mutated genes. However, the feasibility of these approaches for dominantly-inherited diseases-where treatment may require reduction in the expression of a toxic mutant protein resulting from a gain-of-function (GoF) allele-is unclear. Here we show the efficacy of allele-specific RNAi as a potential therapeutic for Charcot-Marie-Tooth type 2D (CMT2D), caused by dominant mutations in glycyl tRNA-synthetase (GARS). A de novo mutation in GARS was identified in a patient with a severe peripheral neuropathy, and a mouse model precisely recreating the mutation was produced. These mice developed a neuropathy by 3-4 weeks-of-age, validating the pathogenicity of the mutation. RNAi sequences targeting mutant GARS mRNA, but not wild-type, were optimized and then packaged into AAV9 for in vivo delivery. This almost completely prevented the neuropathy in mice treated at birth. Delaying treatment until after disease onset showed modest benefit, though this effect decreased the longer treatment was delayed. These outcomes were reproduced in a second mouse model of CMT2D using a vector specifically targeting that allele. The effects were dose dependent, and persisted for at least one year. Our findings demonstrate the feasibility of AAV9-mediated allele-specific knockdown and provide proof-of-concept for gene therapy approaches for dominant neuromuscular diseases.
PMID: 31557132 [PubMed – as supplied by publisher]
PubMed:31557132
Morelli KH, Griffin LB, Pyne NK, Wallace LM, Fowler AM, Oprescu SN, Takase R, Wei N, Meyer-Schuman R, Mellacheruvu D, Kitzman JO, Kocen SG, Hines TJ, Spaulding EL, Lupski JR, Nesvizhskii A, Mancias P, Butler IJ, Yang XL, Hou YM, Antonellis A, Harper SQ, Burgess RW
https://www.ncbi.nlm.nih.gov/pubmed/31525351?dopt=Abstract
https://www.sciencedirect.com/science/article/abs/pii/S000689931930513X?via%3Dihub
Genetic Modifiers and Non-Mendelian Aspects of CMT.
Genetic Modifiers and Non-Mendelian Aspects of CMT.
Brain Res. 2019 Sep 13;:146459
Authors: Bis-Brewer DM, Fazal S, Züchner S
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are amongst the most common inherited diseases in neurology. While great strides have been made to identify the genesis of these diseases, a diagnostic gap of 30-60% remains. Classic models of genetic causation may be limited to fully close this gap and, thus, we review the current state and future role of alternative, non-Mendelian forms of genetics in CMT. Promising synergies exist to further define the full genetic architecture of inherited neuropathies, including affordable whole-genome sequencing, increased data aggregation and clinical collaboration, improved bioinformatics and statistical methodology, and vastly improved computational resources. Given the recent advances in genetic therapies for rare diseases, it becomes a matter of urgency to diagnose CMT patients with great fidelity. Otherwise, they will not be able to benefit from such therapeutic options, or worse, suffer harm when pathogenicity of genetic variation is falsely evaluated. In addition, the newly identified modifier and risk genes may offer alternative targets for pharmacotherapy of inherited and, potentially, even acquired forms of neuropathies.
PMID: 31525351 [PubMed – as supplied by publisher]
PubMed:31525351
Bis-Brewer DM, Fazal S, Züchner S
https://academic.oup.com/hmg/advance-article-abstract/doi/10.1093/hmg/ddz199/5549642?redirectedFrom=fulltext
https://www.ncbi.nlm.nih.gov/pubmed/31411673?dopt=Abstract
Gene replacement therapy after neuropathy onset provides therapeutic benefit in a model of CMT1X.
Gene replacement therapy after neuropathy onset provides therapeutic benefit in a model of CMT1X.
Hum Mol Genet. 2019 Aug 14;:
Authors: Kagiava A, Richter J, Tryfonos C, Karaiskos C, Heslegrave AJ, Sargiannidou I, Rossor AM, Zetterberg H, Reilly MM, Christodoulou C, Kleopa KA
Abstract
X-linked Charcot-Marie-Tooth disease (CMT1X), one of the commonest forms of inherited demyelinating neuropathy, results from GJB1 gene mutations causing loss of function of the gap junction protein connexin32 (Cx32). The aim of this study was to examine whether delayed gene replacement therapy after the onset of peripheral neuropathy can provide a therapeutic benefit in the Gjb1-null/Cx32 knockout (KO) model of CMT1X. After delivery of the LV-Mpz.GJB1 lentiviral vector by a single lumbar intrathecal injection into 6-month old Gjb1-null mice we confirmed expression of Cx32 in lumbar roots and sciatic nerves correctly localized at the paranodal myelin areas. Gjb1-null mice treated with LV-Mpz.GJB1 compared to LV-Mpz.Egfp (mock) vector at the age of 6 months showed improved motor performance at 8 and 10 months. Furthermore, treated mice showed increased sciatic nerve conduction velocities, improvement of myelination and reduced inflammation in lumbar roots and peripheral nerves at 10 months of age, along with enhanced quadriceps muscle innervation. Plasma neurofilament light (NEFL) levels, a clinically relevant biomarker, were also ameliorated in fully treated mice. Intrathecal gene delivery after the onset of peripheral neuropathy offers a significant therapeutic benefit in this disease model, providing a proof of principle for treating patients with CMT1X at different ages.
PMID: 31411673 [PubMed – as supplied by publisher]
PubMed:31411673
Kagiava A, Richter J, Tryfonos C, Karaiskos C, Heslegrave AJ, Sargiannidou I, Rossor AM, Zetterberg H, Reilly MM, Christodoulou C, Kleopa KA
https://www.businesswire.com/news/home/20190718005061/en/Neurogene-Initiates-Natural-History-Study-Charcot-Marie-Tooth
Neurogene Initiates Natural History Study of Charcot-Marie Tooth Disease (CMT4J)
Key data now being collected to support endpoint assessment for future gene therapy clinical trial
https://jnnp.bmj.com/content/early/2019/06/05/jnnp-2019-320717
https://www.ncbi.nlm.nih.gov/pubmed/31167812?dopt=Abstract
Expanding the spectrum of genes responsible for hereditary motor neuropathies.
Related Articles
Expanding the spectrum of genes responsible for hereditary motor neuropathies.
J Neurol Neurosurg Psychiatry. 2019 Jun 05;:
Authors: Previtali SC, Zhao E, Lazarevic D, Pipitone GB, Fabrizi GM, Manganelli F, Mazzeo A, Pareyson D, Schenone A, Taroni F, Vita G, Bellone E, Ferrarini M, Garibaldi M, Magri S, Padua L, Pennisi E, Pisciotta C, Riva N, Scaioli V, Scarlato M, Tozza S, Geroldi A, Jordanova A, Ferrari M, Molineris I, Reilly MM, Comi G, Carrera P, Devoto M, Bolino A
Abstract
BACKGROUND: Inherited peripheral neuropathies (IPNs) represent a broad group of genetically and clinically heterogeneous disorders, including axonal Charcot-Marie-Tooth type 2 (CMT2) and hereditary motor neuropathy (HMN). Approximately 60%-70% of cases with HMN/CMT2 still remain without a genetic diagnosis. Interestingly, mutations in HMN/CMT2 genes may also be responsible for motor neuron disorders or other neuromuscular diseases, suggesting a broad phenotypic spectrum of clinically and genetically related conditions. Thus, it is of paramount importance to identify novel causative variants in HMN/CMT2 patients to better predict clinical outcome and progression.
METHODS: We designed a collaborative study for the identification of variants responsible for HMN/CMT2. We collected 15 HMN/CMT2 families with evidence for autosomal recessive inheritance, who had tested negative for mutations in 94 known IPN genes, who underwent whole-exome sequencing (WES) analyses. Candidate genes identified by WES were sequenced in an additional cohort of 167 familial or sporadic HMN/CMT2 patients using next-generation sequencing (NGS) panel analysis.
RESULTS: Bioinformatic analyses led to the identification of novel or very rare variants in genes, which have not been previously associated with HMN/CMT2 (ARHGEF28, KBTBD13, AGRN and GNE); in genes previously associated with HMN/CMT2 but in combination with different clinical phenotypes (VRK1 and PNKP), and in the SIGMAR1 gene, which has been linked to HMN/CMT2 in only a few cases. These findings were further validated by Sanger sequencing, segregation analyses and functional studies.
CONCLUSIONS: These results demonstrate the broad spectrum of clinical phenotypes that can be associated with a specific disease gene, as well as the complexity of the pathogenesis of neuromuscular disorders.
PMID: 31167812 [PubMed – as supplied by publisher]
PubMed:31167812
Previtali SC, Zhao E, Lazarevic D, Pipitone GB, Fabrizi GM, Manganelli F, Mazzeo A, Pareyson D, Schenone A, Taroni F, Vita G, Bellone E, Ferrarini M, Garibaldi M, Magri S, Padua L, Pennisi E, Pisciotta C, Riva N, Scaioli V, Scarlato M, Tozza S, Geroldi A, Jordanova A, Ferrari M, Molineris I, Reilly MM, Comi G, Carrera P, Devoto M, Bolino A
https://cmtrf.org/blog/new-evidence-for-gene-replacement-in-a-mouse-model-of-charcot-marie-tooth-disease/
https://onlinelibrary.wiley.com/doi/full/10.1002/mgg3.676
https://www.ncbi.nlm.nih.gov/pubmed/31020813?dopt=Abstract
Whole genome sequencing reveals novel IGHMBP2 variant leading to unique cryptic splice-site and Charcot-Marie-Tooth phenotype with early onset symptoms.
Related Articles
Whole genome sequencing reveals novel IGHMBP2 variant leading to unique cryptic splice-site and Charcot-Marie-Tooth phenotype with early onset symptoms.
Mol Genet Genomic Med. 2019 Apr 25;:e00676
Authors: Cassini TA, Duncan L, Rives LC, Newman JH, Phillips JA, Koziura ME, Brault J, Hamid R, Cogan J, Undiagnosed Diseases Network
Abstract
BACKGROUND: Rare variants (RV) in immunoglobulin mu-binding protein 2 (IGHMBP2) [OMIM 600502] can cause an autosomal recessive type of Charcot-Marie-Tooth (CMT) disease [OMIM 616155], an inherited peripheral neuropathy. Over 40 different genes are associated with CMT, with different possible inheritance patterns.
METHODS AND RESULTS: An 11-year-old female with motor delays was found to have distal atrophy, weakness, and areflexia without bulbar or sensory findings. Her clinical evaluation was unrevealing. Whole exome sequencing (WES) revealed a maternally inherited IGHMBP2 RV (c.1730T>C) predicted to be pathogenic, but no variant on the other allele was identified. Deletion and duplication analysis was negative. She was referred to the Undiagnosed Disease Network (UDN) for further evaluation. Whole genome sequencing (WGS) confirmed the previously identified IGHMBP2 RV and identified a paternally inherited non-coding IGHMBP2 RV. This was predicted to activate a cryptic splice site perturbing IGHMBP2 splicing. Reverse transcriptase polymerase chain reaction (RT-PCR) analysis was consistent with activation of the cryptic splice site. The abnormal transcript was shown to undergo nonsense-mediated decay (NMD), resulting in halpoinsufficiency.
CONCLUSION: This case demonstrates the deficiencies of WES and traditional molecular analyses and highlights the advantages of utilization of WGS and functional studies.
PMID: 31020813 [PubMed – as supplied by publisher]
PubMed:31020813
Cassini TA, Duncan L, Rives LC, Newman JH, Phillips JA, Koziura ME, Brault J, Hamid R, Cogan J, Undiagnosed Diseases Network
https://n.neurology.org/content/92/15_Supplement/S58.005
http://indexsmart.mirasmart.com/AAN2019/PDFfiles/AAN2019-001025.pdf
RNAi/gene therapy combined approach as therapeutic strategy for Charcot-Marie-Tooth 2A
https://n.neurology.org/content/92/15_Supplement/P3.4-045
http://indexsmart.mirasmart.com/AAN2019/PDFfiles/AAN2019-003276.pdf
GDAP1-Related Charcot-Marie-Tooth Disease: Additional Evidence for the c.692C>T Variant as a Pathogenic Mutation (P3.4-045)
https://content.iospress.com/articles/journal-of-neuromuscular-diseases/jnd190377
https://www.ncbi.nlm.nih.gov/pubmed/30958311?dopt=Abstract
Modifier Gene Candidates in Charcot-Marie-Tooth Disease Type 1A: A Case-Only Genome-Wide Association Study.
Modifier Gene Candidates in Charcot-Marie-Tooth Disease Type 1A: A Case-Only Genome-Wide Association Study.
J Neuromuscul Dis. 2019 Apr 03;:
Authors: Tao F, Beecham GW, Rebelo AP, Blanton SH, Moran JJ, Lopez-Anido C, Svaren J, Abreu L, Rizzo D, Kirk CA, Wu X, Feely S, Verhamme C, Saporta MA, Herrmann DN, Day JW, Sumner CJ, Lloyd TE, Li J, Yum SW, Taroni F, Baas F, Choi BO, Pareyson D, Scherer SS, Reilly MM, Shy ME, Züchner S, Inherited Neuropathy Consortium
Abstract
BACKGROUND: Charcot-Marie-Tooth disease type 1A (CMT1A) is caused by a uniform 1.5-Mb duplication on chromosome 17p, which includes the PMP22 gene. Patients often present the classic neuropathy phenotype, but also with high clinical variability.
OBJECTIVE: We aimed to identify genetic variants that are potentially associated with specific clinical outcomes in CMT1A.
METHODS: We genotyped over 600,000 genomic markers using DNA samples from 971 CMT1A patients and performed a case-only genome-wide association study (GWAS) to identify potential genetic association in a subset of 644 individuals of European ancestry. A total of 14 clinical outcomes were analyzed in this study.
RESULTS: The analyses yielded suggestive association signals in four clinical outcomes: difficulty with eating utensils (lead SNP rs4713376, chr6 : 30773314, P = 9.91×10-7, odds ratio = 3.288), hearing loss (lead SNP rs7720606, chr5 : 126551732, P = 2.08×10-7, odds ratio = 3.439), decreased ability to feel (lead SNP rs17629990, chr4 : 171224046, P = 1.63×10-7, odds ratio = 0.336), and CMT neuropathy score (lead SNP rs12137595, chr1 : 4094068, P = 1.14×10-7, beta = 3.014).
CONCLUSIONS: While the results require validation in future genetic and functional studies, the detected association signals may point to novel genetic modifiers in CMT1A.
PMID: 30958311 [PubMed – as supplied by publisher]
PubMed:30958311
Tao F, Beecham GW, Rebelo AP, Blanton SH, Moran JJ, Lopez-Anido C, Svaren J, Abreu L, Rizzo D, Kirk CA, Wu X, Feely S, Verhamme C, Saporta MA, Herrmann DN, Day JW, Sumner CJ, Lloyd TE, Li J, Yum SW, Taroni F, Baas F, Choi BO, Pareyson D, Scherer SS, Reilly MM, Shy ME, Züchner S, Inherited Neuropathy Consortium
https://academic.oup.com/brain/advance-article/doi/10.1093/brain/awz064/5419304
https://www.ncbi.nlm.nih.gov/pubmed/30907403?dopt=Abstract
Gene replacement therapy in a model of Charcot-Marie-Tooth 4C neuropathy.
Gene replacement therapy in a model of Charcot-Marie-Tooth 4C neuropathy.
Brain. 2019 Mar 25;:
Authors: Schiza N, Georgiou E, Kagiava A, Médard JJ, Richter J, Tryfonos C, Sargiannidou I, Heslegrave AJ, Rossor AM, Zetterberg H, Reilly MM, Christodoulou C, Chrast R, Kleopa KA
Abstract
Charcot-Marie-Tooth disease type 4C is the most common recessively inherited demyelinating neuropathy that results from loss of function mutations in the SH3TC2 gene. Sh3tc2-/- mice represent a well characterized disease model developing early onset progressive peripheral neuropathy with hypo- and demyelination, slowing of nerve conduction velocities and disturbed nodal architecture. The aim of this project was to develop a gene replacement therapy for treating Charcot-Marie-Tooth disease type 4C to rescue the phenotype of the Sh3tc2-/- mouse model. We generated a lentiviral vector LV-Mpz.SH3TC2.myc to drive expression of the human SH3TC2 cDNA under the control of the Mpz promoter specifically in myelinating Schwann cells. The vector was delivered into 3-week-old Sh3tc2-/- mice by lumbar intrathecal injection and gene expression was assessed 4-8 weeks after injection. Immunofluorescence analysis showed presence of myc-tagged human SH3TC2 in sciatic nerves and lumbar roots in the perinuclear cytoplasm of a subset of Schwann cells, in a dotted pattern co-localizing with physiologically interacting protein Rab11. Quantitative PCR analysis confirmed SH3TC2 mRNA expression in different peripheral nervous system tissues. A treatment trial was initiated in 3 weeks old randomized Sh3tc2-/- littermate mice which received either the full or mock (LV-Mpz.Egfp) vector. Behavioural analysis 8 weeks after injection showed improved motor performance in rotarod and foot grip tests in treated Sh3tc2-/- mice compared to mock vector-treated animals. Moreover, motor nerve conduction velocities were increased in treated Sh3tc2-/- mice. On a structural level, morphological analysis revealed significant improvement in g-ratios, myelin thickness, and ratios of demyelinated fibres in lumbar roots and sciatic nerves of treated Sh3tc2-/- mice. Finally, treated mice also showed improved nodal molecular architecture and reduction of blood neurofilament light levels, a clinically relevant biomarker for axonal injury/degeneration. This study provides a proof of principle for viral gene replacement therapy targeted to Schwann cells to treat Charcot-Marie-Tooth disease type 4C and potentially other similar demyelinating inherited neuropathies.
PMID: 30907403 [PubMed – as supplied by publisher]
PubMed:30907403
Schiza N, Georgiou E, Kagiava A, Médard JJ, Richter J, Tryfonos C, Sargiannidou I, Heslegrave AJ, Rossor AM, Zetterberg H, Reilly MM, Christodoulou C, Chrast R, Kleopa KA
http://www.progettomitofusina2.com/en/malattia_ricerca/Il_nostro_progetto_di_ricerca
TITLE: Genic Therapy for CMT2A: researchers from the Dino Ferrari Center involved in the research thanks to the support of the Mitofusin 2 Project Association.
Our research project aims to develop a possible therapeutic approach for CMT2A, a sensorimotor polyneuropathy, characterized by the death of motor and sensory neurons. The pathology is caused by mutations in the Mitofusin 2 (MFN2) gene, that codes for the MFN2 protein, located in the membrane of a cell organelle, the mitochondria, which represents the power station of cells and performs essential biological functions. As yet, unfortunately, no resolutive therapy is available and very few research groups are involved in studying this disease. Gene therapy represents a promising strategy, since it is designed to correct the genetic cause underlying the disease itself. This type of strategy is giving excellent results in clinical trials with adeno-associated type 9 vectors (AAV9) for the common form of Spinal Muscular Atrophy (SMA), associated with mutations in the SMN gene (www.clinicaltrials.gov).
The clinical trial is currently underway also at the Policlinico of Milan and is followed by our researchers. Therefore, taking advantage of the skills we have acquired in this sector, we propose to use the same type of approach also for the CMT2A. Nevertheless, the causes of this pathology are both the lack of the “healthy” gene, and the presence of the “sick” MFN2 protein are the cause of the pathology. Thus, this aspect of the disease requires, in anticipation of a therapy for the patients, to introduce the “healthy” gene -as it can be done for SMA or for other diseases only caused by the lack of a protein- but also to turn off the sick gene. Furthermore, the MFN2 protein, due to its important role in mitochondria function, must be present “in appropriate quantity”, neither too low nor too high (another aspect that for other proteins isn’t so essential, in relation to their function).
For all these reasons, the development of a gene therapy for CMT2A requires additional efforts in terms of number of experiments and time.
On the bases of these premises, our therapeutic strategy for CMT2A requires to:
- administer two therapeutic vectors: the first one enables to turn off the “sick” gene, the second one allows to express the “healthy” gene. A possible alternative may be the production of a single, larger vector, able to perform both functions.
- obtain optimal MFN2 levels (neither too low nor too high) in order to avoid the appearance of adverse effects.
We have already generated created the necessary constructs, as well as demonstrated the silencing of the “sick” MFN2 gene and the correct expression of the “healthy” MFN2 protein, through the use of the vectors described in point 1, both in cellular and in animal models of this disease. In particular, in the cellular model we observed a marked improvement of the pathological phenotype (Figure 1).
https://clinicaltrials.gov/ct2/show/NCT01193075
Natural History Evaluation of Charcot Marie Tooth Disease (CMT) Types CMT1B, CMT2A, CMT4A, CMT4C, and Others (INC-6601)
ClinicalTrials.gov Identifier: NCT01193075
Recruitment Status : Recruiting
First Posted : September 1, 2010
Last Update Posted : February 20, 2019
See Contacts and Locations
Sponsor:
Michael Shy
Collaborators:
Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta
Johns Hopkins University
National Institute of Neurological Disorders and Stroke (NINDS)
King’s College Hospital NHS Trust
Nemours Children’s Clinic
Stanford University
University of Pennsylvania
University of Rochester
Children’s Hospital of Philadelphia
Sydney Children’s Hospitals Network
Rare Diseases Clinical Research Network
Muscular Dystrophy Association
National Institutes of Health (NIH)
Charcot-Marie-Tooth Association
Massachusetts General Hospital
Cedars-Sinai Medical Center
University of Miami
Dubowitz Neuromuscular Centre
University of Minnesota – Clinical and Translational Science Institute
University of Connecticut
University of Colorado, Denver
Detroit Medical Center
Ohio State University
Information provided by (Responsible Party):
Michael Shy, University of Iowa
Study Details
Study Description
Brief Summary:
This is an observational longitudinal study to determine the natural history and genotype-phenotype correlations of disease causing mutations in Charcot Marie Tooth disease (CMT) type 1B (CMT1B), 2A (CMT2A), 4A (CMT4A), and 4C (CMT4C).
The investigators will also be determine the capability of the newly developed CMT Pediatric Scale (CMT Peds scale) and the Minimal Dataset to measure impairment and perform longitudinal measurements in patients with multiple forms of CMT over a five year window
Condition or disease
Charcot Marie Tooth Disease
Detailed Description:
The Inherited Neuropathies Consortium (INC) is a member of the Rare Disease Clinical Research Network (RDCRN) that is funded by the National Institutes of Health (NIH) through the Office of Rare Diseases (ORD) and the NINDS. The University of Iowa is the lead center in INC which also includes sites from the University of Pennsylvania, Children’s Hospital of Philadelphia (CHOP), the University of Rochester (NY), the National Hospital for Neurology and Neurosurgery in London England, the Dubowicz Neuromuscular Center, and the University of Miami (Florida). The North American CMT Network is an additional consortium that is funded by the Muscular Dystrophy Association (MDA) and the Charcot Marie Tooth Association (CMTA). The University of Iowa is the lead site for this consortium as well. Additional sites include the University of Washington (Seattle), Vanderbilt University, Johns Hopkins University, the University of Sydney, the Besta Neurological Institute in Milan (Italy), Harvard University and the intramural Neurogenetics division of the NIH. All funding sources have agreed to allow us to house data from the two consortia together at the NIH funded Data Management Coordinating Center (DMCC) at the University of South Florida and to group them under the name of the Inherited Neuropathies Consortium (INC).
The inherited peripheral neuropathies are often called Charcot Marie Tooth Disease (CMT), named after the three physician scientists who first described them. These are a heterogenous group of disorders caused by mutations in more than 50 different genes. The diseases cause weakness and loss of sensation in patients. There are no effective treatments for any forms of CMT. There is also limited information on the natural history of how any of the different types of CMT progress and limited outcome measures to measure impairment. The purpose of INC is to investigate the natural history of the different types of CMT, to identify genes that modify the severity of individual types of CMT, to develop and test outcome measures for children with CMT, to develop an interactive website for patients with CMT and to develop the knowledge needed to perform clinical trials in patients with CMT.
To do this, people who have, are suspected to have, or have a family history of an inherited neuropathy will take part in a full-day evaluation. These patients are being seen in order to receive a possible diagnosis of CMT and clinical care, but may also choose to participate in this research project. Most of the testing being performed would be done as part of the standard of care for diagnosing and treating a patient with an inherited neuropathy. Additional testing may be performed on certain subjects if applicable, but the majority of subjects who volunteer for this study are allowing us to use the clinical information obtained during their visit as coded data for natural history purposes and to develop outcome measures.
Because all individuals who attend the CMT clinic would be eligible for participation in this study, when they arrive for their clinic visit, the initial meeting is to review the consent form for the research project. Thus, the evaluation will begin with consenting subjects into the study, if applicable. Then, a variety of tests will take place to measure the presence and severity of symptoms, along with identifying which type of CMT a patient/subject may have. Tests included in this study which are also the standard of care for patients with inherited neuropathies include a neurological evaluation by a trained neurologist, a limited set of nerve conduction studies, physical therapy assessment, orthotist assessment, genetic counseling, and possibly genetic testing to determine the form of CMT. Additional clinical testing which may be used for research includes EMGs or MUNE testing to evaluate possible muscular dystrophies or muscle involvement in the disease. The results from these tests will be used for diagnostic purposes and to give each patient a CMT Neuropathy Score (CMTNS) which is a validated instrument that allows us to assess the severity of disease based on a 36 point scale. If the subject is also involved in the research project, the results will also be used for research purposes.
Additionally, some subjects may undergo testing which is not a part of regular clinical care, but is being done for research purposes only. These tests/procedures may include hand function testing, physical assessment using the CMT Peds Score (an instrument used to evaluate children with CMT – see substudy 6603), research based genetic testing, quality of life questionnaires, or a skin biopsy. All of these tests/procedures are optional, subjects do not have to complete these to be involved in the overall natural history study. Opting out of any of these procedures does not mean that they would be excluded from the study. Some of these procedures would only be performed on certain individuals who are eligible based on age or type of CMT. Skin biopsies are being performed on certain qualified subjects for research purposes in order to grow fibroblast cells to further study various forms of CMT. These individuals would be over 18 and have specific forms of CMT.
Additionally, there are sub-studies that are being performed which SOME subjects may be eligible for. These sub-study options will be discussed with each patient prior to their participation.
Sub-study #6601 – Test-retest protocol addendum for pCMT-QOL. Substudy for a small group of subjects in order to assess the test-retest reliability indicates reproducibility of the pCMT-QOL. In order to calculate test-retest reliability, at a single site of the Inherited Neuropathy Consortium (University of Iowa), 25 subjects aged 8-18 and their parents will be given the appropriate version of pCMT-QOL, as well as the parents of 10 subjects aged 7 and under. These parents will in addition be given a pre-stamped envelope containing the same version of the pCMT-QOL that they took, to be completed by their child and/or parent in 4 weeks and return by mail (in CMT, the disease should not fluctuate in a 4 week interval). Parents will be given one reminder phone call in 6-8 weeks, and the pCMT-QOL forms will be mailed to them again if they are reported missing. Test-retest reliability will be estimated by intraclass correlation coefficients (two-way random effects model) to show correlations between any given individual’s scores in two QOL assessments taken 4 weeks apart.
Sub-study #6602 – This project is to understand modifier genes and how they influence the severity of disease expression, along with identifying new forms of CMT which have not been genetically determined. Subjects who are eligible will either have CMT type 1A (CMT1A) or an unknown form of CMT type 2 (CMT2). Blood will be drawn and sent to the University of Miami where they receive the coded sample and process it through exome sequencing. Subjects will be told that this is optional.
Sub-study #6603 – This project is to develop a new CMT Pediatric Scale (CMT Peds) for Children with CMT. Although there is a validated score (the CMTNS) which measures disease severity for CMT, it is not always applicable to children due to their limited ability to relay information about their symptoms. The CMT Peds scale is being developed and validated in order to measure disease severity in children and have outcome measures available for future clinical trials. Children (defined as 21 and under) being evaluated will be asked to perform functional tasks such as using stairs, walking in a hallway, and performing hand function tests. This information will be used to validate the CMT Peds score. Subjects will be informed that this study is optional.
These tests will be performed in one day during the clinic visit. Patients who attend the clinic are given the option of returning for annual visits to help with disease management. If a patient decides to follow up in the clinic, they will once again be asked to participate in the research. They do not need to participate in the research in order to be seen in the clinic, and they can opt out at any time. If a subject does not follow up by making an appointment in the clinic, they will not be contacted by us to schedule a return visit. All return visits are initiated by the patient/subject. An individual can decide not to participate in the research, but will still be able to be seen in the clinic and receive medical recommendations, treatment, and care by the clinical team who specializes in inherited neuropathies.
https://www.businesswire.com/news/home/20190207005711/en/Neurogene-Announces-Presentation-Preclinical-Data-Gene-Therapies
Neurogene Announces Presentation of Preclinical Data for Gene Therapies to Treat AGU and CMT4J, Two Rare and Devastating Neurodegenerative Disorders
https://cmtrf.org/blog/cmtrf-investigates-a-gene-therapy-program-for-cmt1a/
CMTRF investigates a gene therapy program for CMT1A
https://clinicaltrials.gov/ct2/show/NCT02362438
Intrathecal Administration of scAAV9/JeT-GAN for the Treatment of Giant Axonal Neuropathy
Intrathecal Administration of scAAV9/JeT-GAN for the Treatment of Giant Axonal Neuropathy
The safety and scientific validity of this study is the responsibility of the study sponsor and investigators. Listing a study does not mean it has been evaluated by the U.S. Federal Government. Know the risks and potential benefits of clinical studies and talk to your health care provider before participating. Read our disclaimer for details.
ClinicalTrials.gov Identifier: NCT02362438
Recruitment Status : Recruiting
First Posted : February 13, 2015
Last Update Posted : August 24, 2018
See Contacts and Locations
Sponsor:
National Institute of Neurological Disorders and Stroke (NINDS)
Information provided by (Responsible Party):
National Institutes of Health Clinical Center (CC) ( National Institute of Neurological Disorders and Stroke (NINDS) )
Study Details Tabular ViewNo Results PostedDisclaimerHow to Read a Study Record
Study Description
Go to sections
Brief Summary:
Background:
– The Gigaxonin gene lets the body make a protein chemical called Gigaxonin. Nerves need Gigaxonin to work properly. Giant Axonal Neuropathy (GAN) causes a shortage of functional Gigaxonin. Nerves stop working normally in people with GAN. This causes problems with walking and sometimes with eating, breathing, and many other activities. GAN has no cure. Over time, GAN can shorten a person s life. Researchers want to see if a gene transfer treatment may help people with GAN.
Objectives:
– To see if a gene transfer is safe and shows potential to help people with GAN.
Eligibility:
– People age 5 and older with GAN.
Design:
For 2 months participants must live full-time within 100 miles of the NIH.
Participants will be screened by phone and in person. They will take many tests. Some are listed below. Their medical records will be reviewed. Their caregivers may be contacted.
Participants will have a total of about 30 visits, weekly, monthly, and then yearly over 15 years. They will include many of the tests below.
Physical and nervous system exams.
Blood, urine, and stool samples.
Nerve, lung, heart, and eye tests.
Questionnaires.
MRI scans, nerve biopsies, and spinal taps. Participants will be sedated for some tests.
Speech, memory, muscle, and mobility tests.
Skin biopsy (small sample removed).
Participants will take many medicines. Some require intravenous lines.
Participants will get the gene transfer through an injection by spinal tap into their cerebrospinal fluid, which flows around the brain and spinal cord. The genes are packed in a modified virus that carries the genes to cells in their body. Participants safety is not guaranteed.
Condition or disease Intervention/treatment Phase
Giant Axonal Neuropathy
Gene Transfer
Other: Intrathecal Delivery of scAAV9/JeT-GAN
Drug: Intrathecal Delivery of scAAV9/JeT-GAN
Phase 1
Detailed Description:
This is a non-randomized, phase I, escalating single dose study to assess the safety of the gene transfer vector scAAV9/JeT-GAN through intrathecal delivery to the brain and spinal cord of patients with Giant Axonal Neuropathy (GAN, OMIM No.256850). The primary objective of this study is to assess the safety of the vector following intrathecal delivery in 10-12 GAN patients who are five years of age or older and have mutations which result in positive cross-reactive immunological material (CRIM) status. This terminology is used in other genetic disorders with residual protein expression that would allow for immunotolerance, amenable to enzyme or gene replacement such as in Pompe disease. Mutations which could result in CRIM-positive status include missense mutations, in-frame deletions or duplications or hypomorphic mutations (such as regulatory domain mutations which are leaky such as incomplete splice site mutations). This protocol was amended to include a single GAN patient, 5 years or older CRIM negative patient (‘null mutation patient’). Secondary objectives of this study are 1) to assess motor and sensory disease symptoms pre- and post-treatment, 2) to examine neuropathology in peripheral nerve biopsies in response to treatment, 3) to examine cerebrospinal fluid (CSF) and to conduct CSF studies to assess response to treatment, and 4) to assess vector shedding following vector administration. The first eligible CRIM positive patient will have a genetic diagnosis of giant axonal neuropathy, will be seven years of age or older, and will have a forced vital capacity of greater than or equal to 50 percent predicted value on pulmonary function testing. This study will be the first-in-human trial of intrathecal delivery of scAAV9/JeT-GAN. The primary endpoint will be safety, based upon adverse events and standard laboratory safety evaluations. Secondary endpoints will include clinical and physiological assessment of motor and sensory function, possible rescue of disease pathology in peripheral nerves, examination of CSF in response to treatment, and assessment of vector shedding following administration.
GAN is a chronic neurodegenerative autosomal recessive disease pathologically characterized by enlarged axons with disordered microtubules and intermediate filaments. The disease pathology is due to loss-of-function mutations in the GAN gene, which encodes the protein gigaxonin. Gigaxonin plays a major role in the maintenance of orderly and functional intermediate filament (IF) architecture, which is critical for axonal function. Onset of symptoms, usually at 3-4 years of age, generally manifests with a slightly awkward gait (sensory ataxia). In the peripheral nervous system the disease progressively affects predominantly sensory and motor nerves. By the end of the 2nd decade of life, patients typically are wheelchair dependent with limited use of the arms and little to no use of their legs. During the 2nd decade a tracheostomy or other means of ventilation, as well as a feeding tube, are often necessary. Death normally occurs in the 2nd or 3rd decade of life. There are no statistics on the incidence of the disease, but it is considered extremely rare and there are no effective treatments for the disease. Intrathecal delivery of a gene transfer vector carrying a normal copy of the GAN Gene to the spinal cord and brain offers a potentially effective treatment for GAN.
Study Design
Go to sections
Study Type : Interventional
Estimated Enrollment : 30 participants
Allocation: Non-Randomized
Intervention Model: Sequential Assignment
Masking: None (Open Label)
Primary Purpose: Treatment
Official Title: A Phase I Study of Intrathecal Administration of scAAV9/JeT-GAN for the Treatment of Giant Axonal Neuropathy
Study Start Date : April 24, 2015
Estimated Primary Completion Date : July 1, 2020
Estimated Study Completion Date : April 1, 2030
Resource links provided by the National Library of Medicine
Genetics Home Reference related topics: Giant axonal neuropathy
MedlinePlus related topics: Genes and Gene Therapy
Genetic and Rare Diseases Information Center resources: Giant Axonal Neuropathy Charcot-Marie-Tooth Disease Hereditary Neuropathy With Liability to Pressure Palsies Roussy Levy Syndrome
U.S. FDA Resources
https://academic.oup.com/hmg/article/27/8/1460/4861152
https://www.ncbi.nlm.nih.gov/pubmed/29462293
Intrathecal gene therapy in mouse models expressing CMT1X mutations.
Send to
Hum Mol Genet. 2018 Apr 15;27(8):1460-1473. doi: 10.1093/hmg/ddy056.
Intrathecal gene therapy in mouse models expressing CMT1X mutations.
Kagiava A1, Karaiskos C1, Richter J2, Tryfonos C2, Lapathitis G1, Sargiannidou I1, Christodoulou C2, Kleopa KA1,3.
Author information
Abstract
Gap junction beta-1 (GJB1) gene mutations affecting the gap junction protein connexin32 (Cx32) cause the X-linked Charcot-Marie-Tooth disease (CMT1X), a common inherited neuropathy. Targeted expression of virally delivered Cx32 in Schwann cells following intrathecal injection of lentiviral vectors in the Cx32 knockout (KO) mouse model of the disease has led to morphological and functional improvement. To examine whether this approach could be effective in CMT1X patients expressing different Cx32 mutants, we treated transgenic Cx32 KO mice expressing the T55I, R75W or N175D CMT1X mutations. All three mutants were localized in the perinuclear compartment of myelinating Schwann cells consistent with retention in the ER (T55I) or Golgi (R75W, N175D) and loss of physiological expression in the non-compact myelin. Following intrathecal delivery of the GJB1 gene we detected the virally delivered wild-type (WT) Cx32 in non-compact myelin of T55I KO mice, but only rarely in N175D KO or R75W KO mice, suggesting dominant-negative effects of the R75W and N175D mutants but not of the T55I mutant on co-expressed WT Cx32. GJB1 treated T55I KO mice showed improved motor performance, lower ratios of abnormally myelinated fibers and reduction of inflammatory cells in spinal roots and peripheral nerves compared with mock-treated littermates. Either partial (N175D KO) or no (R75W KO) improvement was observed in the other two mutant lines. Thus, certain CMT1X mutants may interfere with gene addition therapy for CMT1X. Whereas gene addition can be used for non-interfering CMT1X mutations, further studies will be needed to develop treatments for patients harboring interfering mutations.
PMID: 29462293 DOI: 10.1093/hmg/ddy056