Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.745
Filter
1.
J Musculoskelet Neuronal Interact ; 24(2): 228-231, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38826006

ABSTRACT

Increasingly Charcot neuroarthropathy (CN) is being recognized in patients with Charcot-Marie-Tooth (CMT) disease. In this report, we describe a case of CN in a CMT patient, adding to the very scarce literature describing this association. We additionally report his unique evaluation with fluorodeoxyglucose (FDG) and sodium fluoride (NaF) positron emission tomography/computed tomography (PET/CT) scanning, the study of which is limited in CN despite its promising role. A 54-year-old known case of CMT, presented with left foot pain, and swelling for 4 months. Weakness and sensory deficits as a result of CMT were evident in both lower and upper limbs. His x-ray was suggestive of CN. Both FDG and NaF PET/CT scanning demonstrated increased tracer uptake in the first tarsometatarsal joint (TMTJ), in keeping with CN. Recognition of the association of CMT with CN is of vital importance as early diagnosis relies on high clinical suspicion. Characterizing risk factors of CN in CMT patients is still under study. Moreover, there is lack of data evaluating the role of PET/CT in CN and specifically in the context of CMT.


Subject(s)
Charcot-Marie-Tooth Disease , Fluorodeoxyglucose F18 , Positron Emission Tomography Computed Tomography , Sodium Fluoride , Humans , Charcot-Marie-Tooth Disease/diagnostic imaging , Middle Aged , Positron Emission Tomography Computed Tomography/methods , Male , Arthropathy, Neurogenic/diagnostic imaging , Radiopharmaceuticals
2.
Spinal Cord Ser Cases ; 10(1): 38, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796453

ABSTRACT

INTRODUCTION: Charcot Marie tooth disease (CMTD) is also known as Hereditary sensory motor neuropathy. It poses difficulties in attaining intra-operative neuromonitoring signals for deformity correction surgery. In this case report, we intent to mention key points for obtaining good neuromonitoring signals in these cases which increases the safety in scoliosis surgery. CASE PRESENTATION: We present a 14-year-old boy, known case of CMTD, presented with progressive deformity of the back. The child was wheelchair-bound and could walk only a few steps with support. He was unable to maintain a sitting balance without using upper limbs making him functionally quadriparatic. The radiographs showed a double scoliotic curve with costo-pelvic impingement. At the onset, no signals were obtained with routine intra-operative neuromonitoring settings. DISCUSSION: Increasing the sweep length and voltage in our neuro-monitors helped in acquiring the baseline signals and we went ahead to proceed the deformity correction.


Subject(s)
Charcot-Marie-Tooth Disease , Evoked Potentials, Motor , Scoliosis , Humans , Charcot-Marie-Tooth Disease/surgery , Charcot-Marie-Tooth Disease/physiopathology , Male , Adolescent , Scoliosis/surgery , Evoked Potentials, Motor/physiology , Intraoperative Neurophysiological Monitoring/methods
3.
Sci Rep ; 14(1): 10972, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745059

ABSTRACT

Autophagy is a self-degradation system for recycling to maintain homeostasis. p62/sequestosome-1 (p62) is an autophagy receptor that accumulates in neuroglia in neurodegenerative diseases. The objective of this study was to determine the elevation of plasma p62 protein levels in patients with Charcot-Marie-Tooth disease 1A (CMT1A) for its clinical usefulness to assess disease severity. We collected blood samples from 69 CMT1A patients and 59 healthy controls. Plasma concentrations of p62 were analyzed by ELISA, and we compared them with Charcot-Marie-Tooth neuropathy score version 2 (CMTNSv2). A mouse CMT1A model (C22) was employed to determine the source and mechanism of plasma p62 elevation. Plasma p62 was detected in healthy controls with median value of 1978 pg/ml, and the levels were significantly higher in CMT1A (2465 pg/ml, p < 0.001). The elevated plasma p62 levels were correlated with CMTNSv2 (r = 0.621, p < 0.0001), motor nerve conduction velocity (r = - 0.490, p < 0.0001) and disease duration (r = 0.364, p < 0.01). In C22 model, increased p62 expression was observed not only in pathologic Schwann cells but also in plasma. Our findings indicate that plasma p62 measurement could be a valuable tool for evaluating CMT1A severity and Schwann cell pathology.


Subject(s)
Biomarkers , Charcot-Marie-Tooth Disease , Sequestosome-1 Protein , Severity of Illness Index , Charcot-Marie-Tooth Disease/blood , Humans , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/blood , Biomarkers/blood , Male , Female , Animals , Adult , Mice , Middle Aged , Disease Models, Animal , Case-Control Studies , Young Adult , Schwann Cells/metabolism , Schwann Cells/pathology
4.
J Cell Mol Med ; 28(9): e18293, 2024 May.
Article in English | MEDLINE | ID: mdl-38722298

ABSTRACT

Charcot-Marie-Tooth type 2A (CMT2A) is an inherited sensorimotor neuropathy associated with mutations within the Mitofusin 2 (MFN2) gene. These mutations impair normal mitochondrial functioning via different mechanisms, disturbing the equilibrium between mitochondrial fusion and fission, of mitophagy and mitochondrial axonal transport. Although CMT2A disease causes a significant disability, no resolutive treatment for CMT2A patients to date. In this context, reliable experimental models are essential to precisely dissect the molecular mechanisms of disease and to devise effective therapeutic strategies. The most commonly used models are either in vitro or in vivo, and among the latter murine models are by far the most versatile and popular. Here, we critically revised the most relevant literature focused on the experimental models, providing an update on the mammalian models of CMT2A developed to date. We highlighted the different phenotypic, histopathological and molecular characteristics, and their use in translational studies for bringing potential therapies from the bench to the bedside. In addition, we discussed limitations of these models and perspectives for future improvement.


Subject(s)
Charcot-Marie-Tooth Disease , Disease Models, Animal , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/therapy , Charcot-Marie-Tooth Disease/metabolism , Animals , Humans , Mutation , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/pathology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Dynamics/genetics
5.
Sci Adv ; 10(15): eadm7600, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38608019

ABSTRACT

Myelination is essential for neuronal function and health. In peripheral nerves, >100 causative mutations have been identified that cause Charcot-Marie-Tooth disease, a disorder that can affect myelin sheaths. Among these, a number of mutations are related to essential targets of the posttranslational modification neddylation, although how these lead to myelin defects is unclear. Here, we demonstrate that inhibiting neddylation leads to a notable absence of peripheral myelin and axonal loss both in developing and regenerating mouse nerves. Our data indicate that neddylation exerts a global influence on the complex transcriptional and posttranscriptional program by simultaneously regulating the expression and function of multiple essential myelination signals, including the master transcription factor EGR2 and the negative regulators c-Jun and Sox2, and inducing global secondary changes in downstream pathways, including the mTOR and YAP/TAZ signaling pathways. This places neddylation as a critical regulator of myelination and delineates the potential pathogenic mechanisms involved in CMT mutations related to neddylation.


Subject(s)
Charcot-Marie-Tooth Disease , Schwann Cells , Animals , Mice , Myelin Sheath/genetics , Charcot-Marie-Tooth Disease/genetics , Mutation , Protein Processing, Post-Translational
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 443-449, 2024 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-38565510

ABSTRACT

OBJECTIVE: To explore the clinical manifestations and genetic basis for a Chinese pedigree affected with atypical Charcot-Marie-Tooth disease type 1 A (CMT1A). METHODS: A patient admitted to the Department of Neurology, Xijing Hospital Affiliated to Air Force Medical University in June 2022 was selected as the study subject. Clinical data of the patient was collected, and 17 family members from four generations of this pedigree were traced based on pes arcuatus and atypical clinical symptoms. Neuroultrasound and genetic testing were carried out on available family members. Whole exome sequencing and multiple ligation-dependent probe amplification assay were carried out for the proband and some of the affected members of the pedigree. RESULTS: The proband, a 15-year-old male, had presented with paroxystic limb pain with weakness, accompanied by pes cavus and hypertrophy of gastrocnemius muscles, without stork leg sign caused by muscles atrophy in the distal lower extremities. MRI has revealed no sign of fat infiltration in the muscles of both legs. Nerve conduction examination had indicated damages of the sensory and motor nerves of the limbs, mainly with demyelinating changes. Seven members of the pedigree had pes arcuatus, including 5 presenting with paroxysmal neuropathic pain and myasthenia in the limbs, whilst 2 were without any clinical symptoms. Neurosonography of the proband, his brother, father and aunt showed thickened peripheral nerves of the extremities with unclear bundle structure. Genetic analysis revealed a large repeat encompassing exons 1 to 5 of the PMP22 gene and flanking regions (chr17: 15133768_15502298) in some of the affected members, which was predicted to be pathogenic. CONCLUSION: The duplication of PMP22 gene was considered to be pathogenic for this CMT1A pedigree.


Subject(s)
Charcot-Marie-Tooth Disease , Male , Humans , Adolescent , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Pedigree , Myelin Proteins/genetics , Muscle, Skeletal , China , Gene Duplication
7.
Neurology ; 102(9): e209277, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38630962

ABSTRACT

BACKGROUND AND OBJECTIVES: Intramuscular fat fraction (FF) assessed using quantitative MRI (qMRI) has emerged as one of the few responsive outcome measures in CMT1A suitable for future clinical trials. This study aimed to identify the relevance of multiple qMRI biomarkers for tracking longitudinal changes in CMT1A and to assess correlations between MRI metrics and clinical parameters. METHODS: qMRI was performed in CMT1A patients at 2 time points, a year apart, and various metrics were extracted from 3-dimensional volumes of interest at thigh and leg levels. A semiautomated segmentation technique was used, enabling the analysis of central slices and a larger 3D muscle volume. Metrics included proton density (PD), magnetization transfer ratio (MTR), and intramuscular FF. The sciatic and tibial nerves were also assessed. Disease severity was gauged using Charcot Marie Tooth Neurologic Score (CMTNSv2), Charcot Marie Tooth Examination Score, Overall Neuropathy Limitation Scale scores, and Medical Research Council (MRC) muscle strength. RESULTS: Twenty-four patients were included. FF significantly rose in the 3D volume at both thigh (+1.04% ± 2.19%, p = 0.041) and leg (+1.36% ± 1.87%, p = 0.045) levels. The 3D analyses unveiled a length-dependent gradient in FF, ranging from 22.61% ± 10.17% to 26.17% ± 10.79% at the leg level. There was noticeable variance in longitudinal changes between muscles: +3.17% ± 6.86% (p = 0.028) in the tibialis anterior compared with 0.37% ± 4.97% (p = 0.893) in the gastrocnemius medialis. MTR across the entire thigh volume showed a significant decline between the 2 time points -2.75 ± 6.58 (p = 0.049), whereas no significant differences were noted for the 3D muscle volume and PD. No longitudinal changes were observed in any nerve metric. Potent correlations were identified between FF and primary clinical measures: CMTNSv2 (ρ = 0.656; p = 0.001) and MRC in the lower limbs (ρ = -0.877; p < 0.001). DISCUSSION: Our results further support that qMRI is a promising tool for following up longitudinal changes in CMT1A patients, FF being the paramount MRI metric for both thigh and leg regions. It is crucial to scrutinize the postimaging data extraction methods considering that annual changes are minimal (around +1.5%). Given the varied FF distribution, the existence of a length-dependent gradient, and the differential fatty involution across muscles, 3D volume analysis appeared more suitable than single slice analysis.


Subject(s)
Charcot-Marie-Tooth Disease , Humans , Charcot-Marie-Tooth Disease/diagnosis , Muscle, Skeletal , Lower Extremity , Thigh , Magnetic Resonance Imaging/methods
8.
EMBO Mol Med ; 16(5): 1091-1114, 2024 May.
Article in English | MEDLINE | ID: mdl-38589651

ABSTRACT

PAR3/INSC/LGN form an evolutionarily conserved complex required for asymmetric cell division in the developing brain, but its post-developmental function and disease relevance in the peripheral nervous system (PNS) remains unknown. We mapped a new locus for axonal Charcot-Marie-Tooth disease (CMT2) and identified a missense mutation c.209 T > G (p.Met70Arg) in the INSC gene. Modeling the INSCM70R variant in Drosophila, we showed that it caused proprioceptive defects in adult flies, leading to gait defects resembling those in CMT2 patients. Cellularly, PAR3/INSC/LGN dysfunction caused tubulin aggregation and necrotic neurodegeneration, with microtubule-stabilizing agents rescuing both morphological and functional defects of the INSCM70R mutation in the PNS. Our findings underscore the critical role of the PAR3/INSC/LGN machinery in the adult PNS and highlight a potential therapeutic target for INSC-associated CMT2.


Subject(s)
Charcot-Marie-Tooth Disease , Mutation, Missense , Animals , Humans , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Drosophila/genetics , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Disease Models, Animal , Tubulin/genetics , Tubulin/metabolism , Nuclear Proteins , Adaptor Proteins, Signal Transducing
9.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673950

ABSTRACT

Demyelinating Charcot-Marie-Tooth 4G (CMT4G) results from a recessive mutation in the 5'UTR region of the Hexokinase 1 (HK1) gene. HK participates in mitochondrial calcium homeostasis by binding to the Voltage-Dependent Anion Channel (VDAC), through its N-terminal porin-binding domain. Our hypothesis is that CMT4G mutation results in a broken interaction between mutant HK1 and VDAC, disturbing mitochondrial calcium homeostasis. We studied a cohort of 25 CMT4G patients recruited in the French gypsy population. The disease was characterized by a childhood onset, an intermediate demyelinating pattern, and a significant phenotype leading to becoming wheelchair-bound by the fifth decade of life. Co-IP and PLA studies indicated a strong decreased interaction between VDAC and HK1 in the patients' PBMCs and sural nerve. We observed that either wild-type HK1 expression or a peptide comprising the 15 aa of the N-terminal wild-type HK1 administration decreased mitochondrial calcium release in HEK293 cells. However, mutated CMT4G HK1 or the 15 aa of the mutated HK1 was unable to block mitochondrial calcium release. Taken together, these data show that the CMT4G-induced modification of the HK1 N-terminus disrupts HK1-VDAC interaction. This alters mitochondrial calcium buffering that has been shown to be critical for myelin sheath maintenance.


Subject(s)
Calcium , Charcot-Marie-Tooth Disease , Hexokinase , Mitochondria , Voltage-Dependent Anion Channel 1 , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Young Adult , 5' Untranslated Regions/genetics , Calcium/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , HEK293 Cells , Hexokinase/genetics , Hexokinase/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mutation , Protein Binding , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 1/genetics
10.
J Neurol Sci ; 460: 122995, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583391

ABSTRACT

BACKGROUND AND PURPOSE: We previously reported that nerve enlargement assessment by nerve ultrasonography of the intermediate upper limb is applicable for distinguishing demyelinating Charcot-Marie-Tooth disease (CMT) from chronic inflammatory demyelinating polyneuropathy (CIDP). However, differences in the severity and distribution patterns of lower extremity nerve enlargement have not been established for either disease. Therefore, we examined the utility of lower extremity nerve ultrasonography for differentiating between CMT and CIDP. METHODS: Twelve patients with demyelinating CMT and 17 patients with CIDP were evaluated. The median, ulnar, tibial, and fibular nerves were evaluated in three regions: the distal upper extremity, intermediate upper extremity, and lower extremity. Of the 14 selected screening sites, the number of sites that exhibited nerve enlargement (enlargement site number, ESN) in each region was determined. RESULTS: The screening ESNs in the intermediate region and lower extremities were greater in patients with demyelinating CMT than in patients with CIDP and greater than the ESN in the distal region (p = 0.010, p = 0.001, and p = 0.101, respectively). The ESNs in the intermediate region and lower extremities significantly differed among patients with typical CIDP, CIDP variants, and demyelinating CMT (p = 0.084 and p < 0.001). Among the 14 selected screening sites, the combined upper and lower extremity ESNs exhibited the highest AUC (0.92; p < 0.001). CONCLUSIONS: Combining the upper and lower extremities for ultrasonographic nerve measurement more accurately distinguishes CIDP from demyelinating CMT.


Subject(s)
Charcot-Marie-Tooth Disease , Lower Extremity , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Ultrasonography , Humans , Charcot-Marie-Tooth Disease/diagnostic imaging , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnostic imaging , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Male , Female , Middle Aged , Ultrasonography/methods , Adult , Aged , Lower Extremity/diagnostic imaging , Lower Extremity/innervation , Diagnosis, Differential , Peripheral Nerves/diagnostic imaging , Peripheral Nerves/pathology , Young Adult
11.
J Neuromuscul Dis ; 11(3): 625-645, 2024.
Article in English | MEDLINE | ID: mdl-38578900

ABSTRACT

Background: NEFL encodes for the neurofilament light chain protein. Pathogenic variants in NEFL cause demyelinating, axonal and intermediate forms of Charcot-Marie-Tooth disease (CMT) which present with a varying degree of severity and somatic mutations have not been described yet. Currently, 34 different CMT-causing pathogenic variants in NEFL in 174 patients have been reported. Muscular involvement was also described in CMT2E patients mostly as a secondary effect. Also, there are a few descriptions of a primary muscle vulnerability upon pathogenic NEFL variants. Objectives: To expand the current knowledge on the genetic landscape, clinical presentation and muscle involvement in NEFL-related neurological diseases by retrospective case study and literature review. Methods: We applied in-depth phenotyping of new and already reported cases, molecular genetic testing, light-, electron- and Coherent Anti-Stokes Raman Scattering-microscopic studies and proteomic profiling in addition to in silico modelling of NEFL-variants. Results: We report on a boy with a muscular phenotype (weakness, myalgia and cramps, Z-band alterations and mini-cores in some myofibers) associated with the heterozygous p.(Phe104Val) NEFL-variant, which was previously described in a neuropathy case. Skeletal muscle proteomics findings indicated affection of cytoskeletal proteins. Moreover, we report on two further neuropathic patients (16 years old girl and her father) both carrying the heterozygous p.(Pro8Ser) variant, which has been identified as 15% somatic mosaic in the father. While the daughter presented with altered neurophysiology,neurogenic clump feet and gait disturbances, the father showed clinically only feet deformities. As missense variants affecting proline at amino acid position 8 are leading to neuropathic manifestations of different severities, in silico modelling of these different amino acid substitutions indicated variable pathogenic impact correlating with disease onset. Conclusions: Our findings provide new morphological and biochemical insights into the vulnerability of denervated muscle (upon NEFL-associated neuropathy) as well as novel genetic findings expanding the current knowledge on NEFL-related neuromuscular phenotypes and their clinical manifestations. Along this line, our data show that even subtle expression of somatic NEFL variants can lead to neuromuscular symptoms.


Subject(s)
Charcot-Marie-Tooth Disease , Neurofilament Proteins , Phenotype , Humans , Male , Neurofilament Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Retrospective Studies , Child , Adolescent , Female , Mutation
12.
Neurobiol Dis ; 195: 106501, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38583640

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is a genetic peripheral neuropathy caused by mutations in many functionally diverse genes. The aminoacyl-tRNA synthetase (ARS) enzymes, which transfer amino acids to partner tRNAs for protein synthesis, represent the largest protein family genetically linked to CMT aetiology, suggesting pathomechanistic commonalities. Dominant intermediate CMT type C (DI-CMTC) is caused by YARS1 mutations driving a toxic gain-of-function in the encoded tyrosyl-tRNA synthetase (TyrRS), which is mediated by exposure of consensus neomorphic surfaces through conformational changes of the mutant protein. In this study, we first showed that human DI-CMTC-causing TyrRSE196K mis-interacts with the extracellular domain of the BDNF receptor TrkB, an aberrant association we have previously characterised for several mutant glycyl-tRNA synthetases linked to CMT type 2D (CMT2D). We then performed temporal neuromuscular assessments of YarsE196K mice modelling DI-CMT. We determined that YarsE196K homozygotes display a selective, age-dependent impairment in in vivo axonal transport of neurotrophin-containing signalling endosomes, phenocopying CMT2D mice. This impairment is replicated by injection of recombinant TyrRSE196K, but not TyrRSWT, into muscles of wild-type mice. Augmenting BDNF in DI-CMTC muscles, through injection of recombinant protein or muscle-specific gene therapy, resulted in complete axonal transport correction. Therefore, this work identifies a non-cell autonomous pathomechanism common to ARS-related neuropathies, and highlights the potential of boosting BDNF levels in muscles as a therapeutic strategy.


Subject(s)
Axonal Transport , Brain-Derived Neurotrophic Factor , Charcot-Marie-Tooth Disease , Disease Models, Animal , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Mice , Tyrosine-tRNA Ligase/genetics , Tyrosine-tRNA Ligase/metabolism , Humans , Mice, Transgenic , Muscle, Skeletal/metabolism , Receptor, trkB/metabolism , Receptor, trkB/genetics , Mutation
13.
Neurobiol Dis ; 193: 106467, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452947

ABSTRACT

Mutations in the gene encoding MFN2 have been identified as associated with Charcot-Marie-Tooth disease type 2A (CMT2A), a neurological disorder characterized by a broad clinical phenotype involving the entire nervous system. MFN2, a dynamin-like GTPase protein located on the outer mitochondrial membrane, is well-known for its involvement in mitochondrial fusion. Numerous studies have demonstrated its participation in a network crucial for various other mitochondrial functions, including mitophagy, axonal transport, and its controversial role in endoplasmic reticulum (ER)-mitochondria contacts. Considerable progress has been made in the last three decades in elucidating the disease pathogenesis, aided by the generation of animal and cellular models that have been instrumental in studying disease physiology. A review of the literature reveals that, up to now, no definitive pharmacological treatment for any CMT2A variant has been established; nonetheless, recent years have witnessed substantial progress. Many treatment approaches, especially concerning molecular therapy, such as histone deacetylase inhibitors, peptide therapy to increase mitochondrial fusion, the new therapeutic strategies based on MF1/MF2 balance, and SARM1 inhibitors, are currently in preclinical testing. The literature on gene silencing and gene replacement therapies is still limited, except for a recent study by Rizzo et al.(Rizzo et al., 2023), which recently first achieved encouraging results in in vitro and in vivo models of the disease. The near-future goal for these promising therapies is to progress to the stage of clinical translation.


Subject(s)
Charcot-Marie-Tooth Disease , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Charcot-Marie-Tooth Disease/metabolism , Mitochondria/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Phenotype , Mitochondrial Proteins/metabolism , Mutation
14.
Neurology ; 102(7): e209174, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38513194

ABSTRACT

BACKGROUND AND OBJECTIVES: Germline truncating variants in the DRP2 gene (encoding dystrophin-related protein 2) cause the disruption of the periaxin-DRP2-dystroglycan complex and have been linked to Charcot-Marie-Tooth disease. However, the causality and the underlying phenotype of the genetic alterations are not clearly defined. METHODS: This cross-sectional retrospective observational study includes 9 patients with Charcot-Marie-Tooth disease (CMT) with DRP2 germline variants evaluated at 6 centers throughout Spain. RESULTS: We identified 7 Spanish families with 4 different DRP2 likely pathogenic germline variants. In agreement with an X-linked inheritance, men harboring hemizygous DRP2 variants presented with an intermediate form of CMT, whereas heterozygous women were asymptomatic. Symptom onset was variable (36.6 ± 16 years), with lower limb weakness and multimodal sensory loss producing a mild-to-moderate functional impairment. Nerve echography revealed an increase in the cross-sectional area of nerve roots and proximal nerves. Lower limb muscle magnetic resonance imaging confirmed the presence of a length-dependent fatty infiltration. Immunostaining in intradermal nerve fibers demonstrated the absence of DRP2 and electron microscopy revealed abnormal myelin thickness that was also detectable in the sural nerve sections. DISCUSSION: Our findings support the causality of DRP2 pathogenic germline variants in CMT and further define the phenotype as a late-onset sensory and motor length-dependent neuropathy, with intermediate velocities and thickening of proximal nerve segments.


Subject(s)
Charcot-Marie-Tooth Disease , Germ-Line Mutation , Female , Humans , Male , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Myelin Sheath/pathology , Peripheral Nerves/diagnostic imaging , Phenotype , Cross-Sectional Studies , Retrospective Studies , Pedigree , Young Adult , Middle Aged , Aged
15.
J Clin Neuromuscul Dis ; 25(3): 152-156, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38441936

ABSTRACT

ABSTRACT: Hereditary neuropathies are typically associated with an early onset of symptoms, but same types of neuropathies may also manifest late, after the age 50 years. A 62-year-old African American woman presented with a 6-year history of gait unsteadiness and has been using a walker since the age 57 years after an unwitnessed fall. Gradual worsening of walking difficulties was later followed by decreased dexterity. The family history was negative for neuromuscular disorders, including neuropathy. On examination, the patient had both distal and proximal weakness with distal sensory loss to all modalities and hyporeflexia. Charcot Marie Tooth Examination Score was 12. Previous electrodiagnostic testing at the age 60 years showed severe sensorimotor demyelinating polyneuropathy with bilateral severe carpal tunnel syndrome. Genetic testing showed a homozygous pathogenic mutation in SH3TC2 gene (c.2860C>T; p.Arg954*), associated with CMT4C. CMT4C is the most common recessive demyelinating sensorimotor polyneuropathy and overall comprises 0.4%-1.7% of all patients with Charcot-Marie-Tooth disease. It is more common in French Canadians and Spanish Roma and in recent natural history study; only 1 of 56 patients was African American. This report demonstrates sporadic occurrence of CMT4C in other ethnic groups as well.


Subject(s)
Carpal Tunnel Syndrome , Charcot-Marie-Tooth Disease , North American People , Female , Humans , Middle Aged , Black or African American , Charcot-Marie-Tooth Disease/complications , Charcot-Marie-Tooth Disease/genetics
16.
Pediatr Neurol ; 154: 4-8, 2024 May.
Article in English | MEDLINE | ID: mdl-38428336

ABSTRACT

BACKGROUND: Charcot-Marie-Tooth disease (CMT) is a heterogeneous group of inherited peripheral neuropathies. Although the typical disease onset is reported in the second decade, earlier onsets are not uncommon. To date, few studies on pediatric populations have been conducted and the achievement of molecular diagnosis remains challenging. METHODS: During the last 24 years we recruited 223 patients with early-onset hereditary peripheral neuropathies (EOHPN), negative for PMP22 duplication, 72 of them referred by a specialized pediatric hospital. Genetic testing for CMT-associated genes has been carried out with a range of different techniques. RESULTS: Of the 223 EOHPN cases, 43% were classified as CMT1 (demyelinating), 49% as CMT2 (axonal), and 8% as CMTi (intermediate). Genetic diagnosis was reached in 51% of patients, but the diagnostic yield increased to 67% when focusing only on cases from the specialized pediatric neuromuscular centers. Excluding PMP22 rearrangements, no significant difference in diagnostic rate between demyelinating and axonal forms was identified. De novo mutations account for 38% of cases. CONCLUSIONS: This study describes an exhaustive picture of EOHPN in an Italian referral genetic center and analyzes the molecular diagnostic rate of a heterogeneous cohort compared with one referred by a specialized pediatric center. Our data identify MPZ, MFN2, GDAP1, and SH3TC2 genes as the most frequent players in EOHPN. Our study underlines the relevance of a specific neurological pediatric expertise to address the genetic testing and highlights its importance to clarify possible unexpected results when neuropathy is only a secondary clinical sign of a more complex phenotype.


Subject(s)
Charcot-Marie-Tooth Disease , Humans , Child , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/genetics , Genetic Testing , Phenotype , Mutation
17.
Foot Ankle Int ; 45(6): 601-611, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491765

ABSTRACT

BACKGROUND: The complex deformities in cavovarus feet of Charcot-Marie-Tooth (CMT) disease are difficult to evaluate. The aim of this study was to quantify the initial standing alignment correction achieved after joint-sparing CMT cavovarus reconstruction using pre- and postoperative weightbearing computed tomography (WBCT). METHODS: Twenty-nine CMT cavovarus reconstructions were retrospectively analyzed. Three-dimensional measurements were performed using semiautomated software (Bonelogic 2.1) to investigate changes in sagittal, axial, and coronal parameters. Pre- and postoperative data were compared, along with normative data. Correlation among the preoperative measurements and the amount of correction in sagittal, axial, and coronal parameters were analyzed. RESULTS: The sagittal, axial, and coronal malalignment of the hindfoot, and the sagittal and axial malalignment of the forefoot, was significantly improved after corrective surgery (P < .05). Sagittal Meary angle (from 14.8 to 0.1 degrees), axial talonavicular angle (TNA, from 3.6 to 19.2 degrees), and coronal hindfoot alignment (from 11.0 to -11.1 degrees) showed significant changes postoperatively (P < .001). Hindfoot, forefoot sagittal, and forefoot axial parameters reached comparable outcomes compared with normative value (P > .05). Regarding amount of correction, Spearman correlation demonstrated that axial Meary angle and TNA were most strongly related to improvement in sagittal Meary angle and coronal hindfoot alignment. CONCLUSION: Preoperative and postoperative WBCT measurements demonstrated that joint sparing CMT cavovarus reconstruction significantly improved sagittal, axial, and coronal deformities of CMT, and sagittal Meary angle was restored toward normative values. Apparent axial plane correction, the majority of which occurred at the talonavicular joint, had the strongest correlation with deformity correction in multiple planes. This suggests that soft tissue releases and correction of the talonavicular joint may be a key component of a cavovarus foot correction.


Subject(s)
Charcot-Marie-Tooth Disease , Imaging, Three-Dimensional , Tomography, X-Ray Computed , Charcot-Marie-Tooth Disease/surgery , Charcot-Marie-Tooth Disease/diagnostic imaging , Humans , Retrospective Studies , Female , Adult , Male , Talipes Cavus/surgery , Talipes Cavus/diagnostic imaging , Weight-Bearing , Adolescent , Young Adult , Middle Aged , Standing Position
18.
Stem Cell Res ; 77: 103401, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38537501

ABSTRACT

The CMT1A variant accounts for over 60% of cases of Charcot-Marie-Tooth disease (CMT), one of the most common human neuropathies. The cause of CMT1A has been identified as the duplication of PMP22, a myelin protein expressed in Schwann cells. Yet, the pathological mechanisms have not been elucidated, and no treatment is currently available. In our study, we established an iPS cell line from a CMT1A patient with PMP22 duplication. The generated iPSCs maintain pluripotency and in vitro differentiation potency.


Subject(s)
Charcot-Marie-Tooth Disease , Induced Pluripotent Stem Cells , Myelin Proteins , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myelin Proteins/genetics , Myelin Proteins/metabolism , Cell Line , Cell Differentiation , Gene Duplication , Male
19.
Genet Med ; 26(6): 101117, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459834

ABSTRACT

PURPOSE: We describe 3 families with Charcot-Marie-Tooth neuropathy (CMT), harboring a homozygous NDUFS6 NM_004553.6:c.309+5G>A variant previously linked to fatal Leigh syndrome. We aimed to characterize clinically and molecularly the newly identified patients and understand the mechanism underlying their milder phenotype. METHODS: The patients underwent extensive clinical examinations. Exome sequencing was done in 4 affected individuals. The functional effect of the c.309+5G>A variant was investigated in patient-derived EBV-transformed lymphoblasts at the complementary DNA, protein, and mitochondrial level. Alternative splicing was evaluated using complementary DNA long-read sequencing. RESULTS: All patients presented with early-onset, slowly progressive axonal CMT, and nystagmus; some exhibited additional central nervous system symptoms. The c.309+5G>A substitution caused the expression of aberrantly spliced transcripts and negligible levels of the canonical transcript. Immunoblotting showed reduced levels of mutant isoforms. No detectable defects in mitochondrial complex stability or bioenergetics were found. CONCLUSION: We expand the clinical spectrum of NDUFS6-related mitochondrial disorders to include axonal CMT, emphasizing the clinical and pathophysiologic overlap between these 2 clinical entities. This work demonstrates the critical role that alternative splicing may play in modulating the severity of a genetic disorder, emphasizing the need for careful consideration when interpreting splice variants and their implications on disease prognosis.


Subject(s)
Alternative Splicing , Charcot-Marie-Tooth Disease , Mitochondrial Diseases , Humans , Alternative Splicing/genetics , Male , Female , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Child , NADH Dehydrogenase/genetics , Pedigree , Mutation/genetics , Phenotype , Exome Sequencing , Leigh Disease/genetics , Leigh Disease/pathology , Mitochondria/genetics , Mitochondria/pathology , Electron Transport Complex I/genetics , Adult , Child, Preschool , Adolescent
20.
J Neurol ; 271(6): 3546-3553, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38549004

ABSTRACT

BACKGROUND: Peripheral neuropathies in mitochondrial disease are caused by mutations in nuclear genes encoding mitochondrial proteins, or in the mitochondrial genome. Whole exome or genome sequencing enable parallel testing of nuclear and mtDNA genes, and it has significantly advanced the genetic diagnosis of inherited diseases. Despite this, approximately 40% of all Charcot-Marie-Tooth (CMT) cases remain undiagnosed. METHODS: The genome-phenome analysis platform (GPAP) in RD-Connect was utilised to create a cohort of 2087 patients with at least one Human Phenotype Ontology (HPO) term suggestive of a peripheral neuropathy, from a total of 10,935 patients. These patients' genetic data were then analysed and searched for variants in known mitochondrial disease genes. RESULTS: A total of 1,379 rare variants were identified, 44 of which were included in this study as either reported pathogenic or likely causative in 42 patients from 36 families. The most common genes found to be likely causative for an autosomal dominant neuropathy were GDAP1 and GARS1. We also detected heterozygous likely pathogenic variants in DNA2, MFN2, DNM2, PDHA1, SDHA, and UCHL1. Biallelic variants in SACS, SPG7, GDAP1, C12orf65, UCHL1, NDUFS6, ETFDH and DARS2 and variants in the mitochondrial DNA (mtDNA)-encoded MT-ATP6 and MT-TK were also causative for mitochondrial CMT. Only 50% of these variants were already reported as solved in GPAP. CONCLUSION: Variants in mitochondrial disease genes are frequent in patients with inherited peripheral neuropathies. Due to the clinical overlap between mitochondrial disease and CMT, agnostic exome or genome sequencing have better diagnostic yields than targeted gene panels.


Subject(s)
Mitochondrial Diseases , Peripheral Nervous System Diseases , Humans , Male , Female , Peripheral Nervous System Diseases/genetics , Adult , Mitochondrial Diseases/genetics , Middle Aged , Aged , Young Adult , Mutation , Mitochondrial Proteins/genetics , Cohort Studies , Adolescent , Charcot-Marie-Tooth Disease/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...